US20100092526A1 - Nanoemulsion therapeutic compositions and methods of using the same - Google Patents

Nanoemulsion therapeutic compositions and methods of using the same Download PDF

Info

Publication number
US20100092526A1
US20100092526A1 US12/567,470 US56747009A US2010092526A1 US 20100092526 A1 US20100092526 A1 US 20100092526A1 US 56747009 A US56747009 A US 56747009A US 2010092526 A1 US2010092526 A1 US 2010092526A1
Authority
US
United States
Prior art keywords
oil
nanoemulsion
sodium
ammonium chloride
acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/567,470
Inventor
James R. Baker, Jr.
Tarek Hamouda
Joyce A. Sutcliffe
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanobio Corp
Original Assignee
Nanobio Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanobio Corp filed Critical Nanobio Corp
Priority to US12/567,470 priority Critical patent/US20100092526A1/en
Assigned to NANOBIO CORPORATION reassignment NANOBIO CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAKER, JR., JAMES R., HAMOUDA, TAREK, SUTCLIFFE, JOYCE A.
Publication of US20100092526A1 publication Critical patent/US20100092526A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/047Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates having two or more hydroxy groups, e.g. sorbitol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/14Quaternary ammonium compounds, e.g. edrophonium, choline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4425Pyridinium derivatives, e.g. pralidoxime, pyridostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • the present invention relates to therapeutic nanoemulsion compositions and to methods of utilizing the same.
  • nanoemulsion compositions are described herein that find use in the treatment and/or prevention of infection (e.g., respiratory infection (e.g., associated with cystic fibrosis)), in burn wound management, and in immunogenic compositions (e.g., comprising a Burkholderia antigen) that generate an effective immune response (e.g., against a bacterial species of the genus Burkholderia ) in a subject administered the immunogenic composition.
  • infections e.g., respiratory infection (e.g., associated with cystic fibrosis)
  • immunogenic compositions e.g., comprising a Burkholderia antigen
  • an effective immune response e.g., against a bacterial species of the genus Burkholderia
  • Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine), industrial, and research applications.
  • Bacterial infection caused by opportunistic and/or pathogenic bacteria e.g., Burkholderia cepacia, Staphylococcus aureus and Pseudomonas aeruginosa
  • opportunistic and/or pathogenic bacteria e.g., Burkholderia cepacia, Staphylococcus aureus and Pseudomonas aeruginosa
  • certain types of individuals are prone to respiratory infection (e.g., by bacteria (e.g., opportunistic bacteria), viruses, fungi and/or parasites) including the immunocompromised, elderly, cancer chemotherapy patients, individuals suffering from asthma, individual suffering from genetically inherited disease (e.g., cystic fibrosis) and virally infected individuals (e.g., infected with influenza virus, respiratory syncytial virus (RSV), adenovirus and/or human immunodeficiency virus).
  • RSV respiratory syncytial virus
  • wounds e.g., burn wounds
  • present on a subject provide an ideal location for bacterial growth and survival.
  • bacterial infection e.g., bacteremia
  • the present invention is directed to a method of treating or preventing a respiratory infection in a subject having Cystic fibrosis (CF).
  • the method comprises administering a nanoemulsion to the subject, wherein the subject is susceptible to or has an infection by one or more gram-positive or gram-negative bacterial species.
  • the nanoemulsion comprises water, at least one organic solvent, at least one surfactant, and at least one oil.
  • the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • the respiratory infection is associated with a bacterial biofilm present in the lungs of the subject.
  • a method of treating or preventing an infection in a subject having a burn wound comprises administering a nanoemulsion to the subject, wherein the subject is susceptible to or has an infection by one or more gram-positive or gram-negative bacterial species.
  • the nanoemulsion comprises water, at least one organic solvent, at least one surfactant, and at least one oil.
  • the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • the bacteria can be any known gram-negative or gram-positive bacteria.
  • the bacterial species is selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp.
  • the bacterial species can also be selected from the group consisting of Pseudomonas aeruginosa, B. cenocepacia, A. baumannii, Stenotrophomonas maltophilia, Staphylococcus aureus, H. influenzae, E. coli, K. pneumoniae , and Proteus mirabilis.
  • a method of treating or preventing an Haemophilus influenzae infection in a non-CF subject comprises administering a nanoemulsion to the subject having or at risk of having a Haemophilus influenzae infection.
  • the nanoemulsion comprises water, at least one organic solvent, at least one surfactant, and at least one oil.
  • the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • All methods described herein may further comprise administering one or more antibiotics either before, during, or after administration of the nanoemulsion.
  • the administration of a nanoemulsion and at least one antibiotic is synergistic.
  • the “synergy” may be as defined by a fractional inhibitory concentration (FIC) index, a fractional bactericidal concentration (FBC) index, or a combination thereof.
  • FBC fractional inhibitory concentration
  • Methods comprising administering an antibiotic encompass the use of any antibiotic.
  • Exemplary antibiotics include, but are not limited to, a polymyxin antibiotic (e.g. colistin) or any aminoglycoside (e.g. tobramycin).
  • the nanoemulsion does not exhibit any antagonism with the antibiotic.
  • the nanoemulsion exhibits minimal or no toxicity or side effects. In addition, preferably the nanoemulsion does not exhibit resistance.
  • the minimum inhibitory concentration (MIC), the minimum bactericidal concentration (MBC), or a combination thereof for the nanoemulsion demonstrate bacteriostatic or bactericidal activity for the nanoemulsion.
  • one or more bacterial species may exhibit resistance against one or more antibiotics.
  • the bacterial species can be methicillin-resistant Staphylococcus aureus (MRSA).
  • the nanoemulsion is not absorbed systemically in the human subject, or very little of the nanoemulsion is absorbed systemically in the human subject.
  • the nanoemulsion can be delivered using any pharmaceutically acceptable means.
  • the nanoemulsion can be delivered via inhalation, topically, topically to a mucosal surface, via nebulization, or via any combination thereof.
  • compositions useful in the methods of the invention include, but are not limited to, nanoemulsions comprising: (a) water; (b) ethanol or glycerol; (c) cetylpyridinium chloride (CPC), or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824); (d) soybean oil; and (e) Poloxamer 407, Tween 80, or Tween 20.
  • the nanoemulsion can additionally comprise EDTA.
  • FIG. 1 shows killing of Burkholderia cepacia in PBS by 20% Nanemulsion (NE) alone or with 20 mM EDTA in 10 minutes.
  • FIG. 2 shows killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 20 minutes.
  • FIG. 3 shows killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 40 minutes.
  • FIG. 4 shows (A) killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 40 minutes; (B) killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 60 minutes; and (C) killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 60 minutes.
  • FIG. 5 shows killing of B. cepacia in hypertonic saline (6% NaCl) at 15 and 30 Minutes by NE alone and NE with EDTA.
  • FIG. 6 shows killing of B. cepacia and P. aeruginosa in 7% NaCl within 15 min.
  • FIG. 7 shows killing of B. cepacia and P. aeruginosa (Mixed-culture) in 7% NaCl within 15 minutes.
  • FIG. 8 shows a microtiter serial dilution MIC assay. Numbers above the plate indicate the concentration ( ⁇ g/ml CPC) of P 407 5EC added to each column of wells. The bacterial species added to wells in each row is indicated on the left. The MICs for these strains (top to bottom) are ⁇ 15.6 ⁇ g/ml 125 ⁇ g/ml, and 31.2 ⁇ g/ml.
  • FIG. 9 shows distribution of P 407 5EC MICs for 150 Burkholderia and non- Burkholderia strains
  • FIG. 10 shows a time-kill study of P 407 5EC activity and concentration-dependent inhibition of P 407 5EC activity by CF sputum.
  • A 10 5 CFU/ml of B. multivorans ATCC 17616 were exposed to various concentrations of P 407 5EC around the MIC (62.5 ⁇ g/ml) and viable bacterial counts were determined at the times indicated.
  • B 10 5 CFU/ml of B. multivorans ATCC 17616 were exposed to various concentrations of P 407 5EC in the presence of three concentrations of CF sputum for 24 hr before viable bacterial counts were determined. Numbers and symbols in inset boxes indicate concentrations of P 407 5EC ( ⁇ g/ml CPC) used.
  • FIG. 11 shows in vitro activity of P 407 5EC.
  • FIG. 12 shows in vitro activity of P 407 5EC against biofilm bacteria and in the presence of CF sputum.
  • FIG. 13 shows influence of P 407 5EC on B. cepacia, P. aeruginosa and E. coli individual biofilms.
  • FIG. 14 shows influence of P 407 5EC on mixed biofilms.
  • FIG. 15 shows influence of P 407 5EC on mixed biofilms.
  • FIG. 16 shows influence of P 407 5EC on escape colonies of B. cepacia biofilms.
  • FIG. 17 shows serial, two-fold dilutions of P 407 5EC in one embodiment of the invention.
  • FIG. 18 shows age specific prevalence of respiratory infections in CF patients.
  • FIG. 19A shows a Fractional Inhibitory Concentration Index
  • FIG. 19B shows a graph demonstrating the relationship between antagonism, indifference, and synergy for Antimicrobial A (micrograms/mL) and Antimicrobial B (micrograms/mL);
  • FIGS. 20A-D show figures of electron microscopy.
  • FIG. 2A shows 0 min, no treatment control;
  • FIG. 2B shows 10 minutes after treatment;
  • FIG. 2C shows 20 minutes after treatment; and
  • FIG. 2D shows 30 minutes after treatment.
  • FIG. 21 shows topical application of nanoemulsion reduces P. aeruginosa growth in burn wounds.
  • Male Sprague-Dawley rats received partial thickness burn wounds.
  • animals were inoculated with 10 6 CFU P. aeruginosa .
  • the animals were treated with topical saline (Control), placebo (W 20 5GBA 2 ED without benzalkonium chloride), W 20 5GBA 2 ED (nanoemulsion), or Sulfamylon (mafenide acetate).
  • mice were sacrificed, skin samples obtained and homogenized, plated, and CFUs counted.
  • the scatter plot represents cultured CFUs for each individual animal.
  • the median value for each group is plotted as a horizontal line. There was minimal pathogen growth in 12 out of 23 of the NB-201 treated animals. A majority of the control (29/32) and placebo (9/12) animals with burn injury had evidence of wound infection based on a positive quantitative wound culture with significantly more bacteria present in the wound than those animals treated with W 20 5GBA 2 ED. p ⁇ 0.0001, Kruskal-Wallis test, p ⁇ 0.05 for saline vs. W 20 5GBA 2 ED, placebo vs. W 20 5GBA 2 ED, and saline vs. Sulfamylon, Dunn's multiple comparison test.
  • FIG. 22 shows nanoemulsion treatment following burn injury attenuates dermal proinflammatory cytokine expression.
  • Burn injured animals treated with nanoemulsion had decreased levels of IL-1 ⁇ (1773 ⁇ 516 vs. 5625 ⁇ 1743 pg/mL) and CINC-3 (225 ⁇ 66 vs. 1589 ⁇ 527 pg/mL) when compared to untreated partial thickness burned animals.
  • nanoemulsion and Sulfamylon treatment groups demonstrated reduced dermal neutrophil sequestration when compared to saline and placebo as evidenced on myeloperoxidase assay (W 20 5GBA 2 ED: 0.09 ⁇ 0.02, Sulfamylon: 0.08 ⁇ 0.02, Saline: 0.40 ⁇ 0.06, Placebo: 0.45 ⁇ 0.11 ⁇ g/mL). *p ⁇ 0.05, Tukey's multiple comparison test.
  • FIG. 24 shows burn injury upregulates dermal TGF- ⁇ expression and treatment with nanoemulsion in the setting of burn wound infection decreases the level of TGF- ⁇ in the wound.
  • Dermal levels of the anti-inflammatory cytokines IL-10 and TGF- ⁇ were measured in the burn wound 32 hours after thermal injury in animals treated with nanoemulsion, and in animals exposed to bacteria and treated with nanoemulsion or Sulfamylon.
  • FIG. 25 shows cross sectional histology of burn skin following infection with P. aeruginosa and treatment with saline or nanoemulsion. Both views of skin are 32 hours after burn injury.
  • FIG. 26 shows evans blue assay to quantify capillary leak and tissue edema.
  • Evans blue is a dye that binds to serum albumin and can be quantitated to determine vascular permeability.
  • FIG. 27 shows photomicrographs of partial thickness burned skin with fluorescence labeled TUNEL staining to detect hair follicle cell apoptosis. Skin samples were harvested at 12 hours post-burn. Treatment was performed at 0 and 8 hours following thermal injury. All images are at 40 ⁇ magnification.
  • FIG. 28 shows TUNEL assay for hair follicle cell apoptosis following a partial thickness burn injury.
  • Skin samples from sham, burn+saline (control), burn+placebo (W 20 5GBA 2 ED without benzalkonium chloride), and burn+W 20 5GBA 2 ED were sectioned for fluorescein-labeled TUNEL assay.
  • Treatment was performed at time 0 and 8 hours post injury. No bacterial inoculation of the burned skin was done in this experiment.
  • There was a significant reduction in the degree of hair follicle apoptosis among burn injured and treated animals for tissue samples harvested at 12 hours post injury (p 0.006, 1-way ANOVA). Differences were found for sham vs. burn+saline, burn+saline vs. burn+placebo, and burn+saline vs. burn+W 20 5GBA 2 ED (*p ⁇ 0.05, Tukey's multiple comparison test).
  • FIG. 29 shows characterization of the OMP preparation.
  • FIG. 30 shows antibody responses against B. cenocepacia OMP.
  • FIG. 31 shows type of cellular immune responses induced by nasal OMP-NE vaccine.
  • OMP-NE NE
  • OMP-PBS PBS
  • FIG. 32 shows identification of protein epitopes and LPS in OMP preparations.
  • Lane 1. Protein ladder; Lane 2. Crude OMP preparation; Lane 3. Proteinase K digested crude OMP preparation; Lane 4. Endotoxin-depleted OMP; Lane 5. Proteinase K digested endotoxin-depleted OMP.
  • Western blots were probed with serum from either OMP in PBS or OMP-NE immunized mice or with a monoclonal anti- Pseudomonas mallei LPS antibody as indicated.
  • FIG. 34 shows pulmonary and splenic colonization assay.
  • microorganism refers to any species or type of microorganism, including but not limited to, bacteria, viruses, archaea, fungi, protozoans, mycoplasma, prions, and parasitic organisms.
  • the term microorganism encompasses both those organisms that are in and of themselves pathogenic to another organism (e.g., animals, including humans, and plants) and those organisms that produce agents that are pathogenic to another organism, while the organism itself is not directly pathogenic or infective to the other organism.
  • pathogen refers a biological agent that causes a disease state (e.g., infection, sepsis, etc.) in a host.
  • pathogens include, but are not limited to, viruses, bacteria, archaea, fungi, protozoans, mycoplasma, prions, and parasitic organisms.
  • bacteria and “bacterium” refer to all prokaryotic organisms, including those within all of the phyla in the Kingdom Procaryotae. It is intended that the term encompass all microorganisms considered to be bacteria including Mycoplasma, Chlamydia, Actinomyces, Streptomyces , and Rickettsia . All forms of bacteria are included within this definition including cocci, bacilli, spirochetes, spheroplasts, protoplasts, etc. Also included within this term are prokaryotic organisms that are Gram-negative or Gram-positive. “Gram-negative” and “Gram-positive” refer to staining patterns with the Gram-staining process, which is well known in the art.
  • Gram-positive bacteria are bacteria that retain the primary dye used in the Gram stain, causing the stained cells to generally appear dark blue to purple under the microscope.
  • Gram-negative bacteria do not retain the primary dye used in the Gram stain, but are stained by the counterstain. Thus, Gram-negative bacteria generally appear red.
  • bacteria are continuously cultured.
  • bacteria are uncultured and existing in their natural environment (e.g., at the site of a wound or infection) or obtained from patient tissues (e.g., via a biopsy). Bacteria may exhibit pathological growth or proliferation.
  • bacteria examples include, but are not limited to, bacterial cells of a genus of bacteria selected from the group comprising Salmonella, Shigella, Escherichia, Enterobacter, Serratia, Proteus, Yersinia, Citrobacter, Edwardsiella, Providencia, Klebsiella, Hafnia, Ewingella, Kluyvera, Morganella, Planococcus, Stomatococcus, Micrococcus, Staphylococcus, Vibrio, Aeromonas, Plessiomonas, Haemophilus, Actinobacillus, Pasteurella, Mycoplasma, Ureaplasma, Rickettsia, Coxiella, Rochalimaea, Ehrlichia, Streptococcus, Enterococcus, Aerococcus, Gemella, Lactococcus, Leuconostoc, Pedicoccus, Bacillus, Corynebacterium, Arcanobacterium, Actinomyces, Rho
  • microorganism and “microbe” refer to any species or type of microorganism, including but not limited to, bacteria, archaea, fungi, protozoans, mycoplasma, and parasitic organisms.
  • fungi is used in reference to eukaryotic organisms such as molds and yeasts, including dimorphic fungi.
  • disease and “pathologic condition” are used interchangeably, unless indicated otherwise herein, to describe a deviation from the condition regarded as normal or average for members of a species or group (e.g., humans), and which is detrimental to an affected individual under conditions that are not inimical to the majority of individuals of that species or group.
  • a deviation can manifest as a state, signs, and/or symptoms (e.g., diarrhea, nausea, fever, pain, blisters, boils, rash, immune suppression, inflammation, etc.) that are associated with any impairment of the normal state of a subject or of any of its organs or tissues that interrupts or modifies the performance of normal functions.
  • a disease or pathological condition may be caused by or result from contact with a microorganism (e.g., a pathogen or other infective agent (e.g., a virus or bacteria)), may be responsive to environmental factors (e.g., malnutrition, industrial hazards, and/or climate), may be responsive to an inherent defect of the organism (e.g., genetic anomalies) or to combinations of these and other factors.
  • a microorganism e.g., a pathogen or other infective agent (e.g., a virus or bacteria)
  • environmental factors e.g., malnutrition, industrial hazards, and/or climate
  • an inherent defect of the organism e.g., genetic anomalies
  • Respiratory and “respiration” refer to the process by which oxygen is taken into the body and carbon dioxide is discharged, through the bodily system including the nose, throat, larynx, trachea, bronchi and lungs.
  • “Respiratory infection” and “pulmonary infection” refer to an infection (e.g., bacterial, viral, fungal, etc.) of the respiratory tract.
  • the respiratory tract comprises the upper respiratory tract (e.g., nose, throat or pharynx, and larynx); the airways (e.g.,: voice box or larynx, windpipe or trachea, and bronchi); and the lungs (e.g., bronchi, bronchioles, alveolar ducts, alveolar sacs, and alveoli).
  • Respiratory disease refers to any one of several ailments that involve inflammation and affect a component of the respiratory system including especially the trachea, bronchi and lungs.
  • obstructive respiratory disease e.g., asthma; bronchitis; and chronic obstructive pulmonary disease, referred to as COPD
  • upper airway disease e.g., such as otitis media, and rhinitis/sinusitis
  • insterstitial lung disease e.g., pneumonia, pneyumocystis carinii, and respiratory syncitial virus (RSV)
  • RSV respiratory syncitial virus
  • ALI acute lung injury
  • ARDS acute respiratory distress syndrome
  • MAS meconium aspiration syndrome
  • RDS respiratory distress syndrome
  • ALI is associated with conditions that either directly or indirectly injure the air sacs of the lung, the alveoli.
  • ALI is a syndrome of inflammation and increased permeability of the lungs with an associated breakdown of the lungs' surfactant layer.
  • ARDS The most serious manifestation of ALI is ARDS.
  • VILI mechanical ventilator induced lung injury
  • smoke inhalation pneumonia
  • sepsis sepsis.
  • subject refers to organisms to be treated by the compositions of the present invention. Such organisms include animals (domesticated animal species, wild animals), and humans.
  • the terms “inactivating,” “inactivation” and grammatical equivalents, when used in reference to a microorganism refer to the killing, elimination, neutralization and/or reducing the capacity of the microorganism to infect and/or cause a pathological response and/or disease in a host.
  • fusigenic is intended to refer to an emulsion that is capable of fusing with the membrane of a microbial agent (e.g., a bacterium or bacterial spore). Specific examples of fusigenic emulsions are described herein.
  • the term “lysogenic” refers to an emulsion (e.g., a nanoemulsion) that is capable of disrupting the membrane of a microbial agent (e.g., a virus (e.g., viral envelope) or a bacterium, bacterial spore, or bacterial biofilm).
  • a microbial agent e.g., a virus (e.g., viral envelope) or a bacterium, bacterial spore, or bacterial biofilm.
  • a lysogenic and a fusigenic agent in the same composition produces an enhanced inactivating effect compared to either agent alone. Methods and compositions using this improved antimicrobial composition are described in detail herein.
  • nanoemulsion includes dispersions or droplets, as well as other lipid structures that can form as a result of hydrophobic forces that drive apolar residues (i.e., long hydrocarbon chains) away from water and drive polar head groups toward water, when a water immiscible oily phase is mixed with an aqueous phase.
  • lipid structures include, but are not limited to, unilamellar, paucilamellar, and multilamellar lipid vesicles, micelles, and lamellar phases.
  • the terms “contact,” “contacted,” “expose,” and “exposed,” when used in reference to a nanoemulsion and a live microorganism refer to bringing one or more nanoemulsions into contact with a microorganism (e.g., a pathogen) such that the nanoemulsion kill and/or attenuate growth of the microorganism or pathogenic agent, if present.
  • a microorganism e.g., a pathogen
  • the present invention is not limited by the amount or type of nanoemulsion used for microorganism killing and/or growth attenuation.
  • a variety of nanoemulsion that find use in the present invention are described herein and elsewhere (e.g., nanoemulsions described in U.S. Pat. Apps.
  • Ratios and amounts of nanoemulsion are contemplated in the present invention including, but not limited to, those described herein (e.g., in Examples 1-4, the Figures associated therewith and FIG. 17 ).
  • surfactant refers to any molecule having both a polar head group, which energetically prefers solvation by water, and a hydrophobic tail which is not well solvated by water.
  • cationic surfactant refers to a surfactant with a cationic head group.
  • anionic surfactant refers to a surfactant with an anionic head group.
  • HLB Index Number refers to an index for correlating the chemical structure of surfactant molecules with their surface activity.
  • the HLB Index Number may be calculated by a variety of empirical formulas as described by Meyers, (Meyers, Surfactant Science and Technology, VCH Publishers Inc., New York, pp. 231-245 [1992]), incorporated herein by reference.
  • the HLB Index Number of a surfactant is the HLB Index Number assigned to that surfactant in McCutcheon's Volume 1: Emulsifiers and Detergents North American Edition, 1996 (incorporated herein by reference).
  • the HLB Index Number ranges from 0 to about 70 or more for commercial surfactants. Hydrophilic surfactants with high solubility in water and solubilizing properties are at the high end of the scale, while surfactants with low solubility in water which are good solubilizers of water in oils are at the low end of the scale.
  • interaction enhancers refers to compounds that act to enhance the interaction of an emulsion with a microorganism (e.g., with a cell wall of a bacteria (e.g., a Gram negative bacteria) or with a viral envelope.
  • Contemplated interaction enhancers include, but are not limited to, chelating agents (e.g., ethylenediaminetetraacetic acid (EDTA), ethylenebis(oxyethylenenitrilo)tetraacetic acid (EGTA), and the like) and certain biological agents (e.g., bovine serum abulmin (BSA) and the like).
  • buffer or “buffering agents” refer to materials which when added to a solution, cause the solution to resist changes in pH.
  • reducing agent and “electron donor” refer to a material that donates electrons to a second material to reduce the oxidation state of one or more of the second material's atoms.
  • monovalent salt refers to any salt in which the metal (e.g., Na, K, or Li) has a net 1+ charge in solution (i.e., one more proton than electron).
  • chelator or “chelating agent” refer to any materials having more than one atom with a lone pair of electrons that are available to bond to a metal ion.
  • solution refers to an aqueous or non-aqueous mixture.
  • the term “effective amount” refers to the amount of a composition (e.g., a composition comprising a nanoemulsion) sufficient to effect a beneficial or desired result (e.g., to treat and/or prevent infection (e.g., through bacterial cell killing and/or prevention of bacterial cell growth).
  • a beneficial or desired result e.g., to treat and/or prevent infection (e.g., through bacterial cell killing and/or prevention of bacterial cell growth).
  • An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • a composition for inducing an immune response refers to a composition that, when administered to a subject (e.g., once, twice, three times or more (e.g., separated by weeks, months or years)), stimulates, generates and/or elicits an immune response in the subject (e.g., resulting in total or partial immunity to a microorganism (e.g., pathogen) capable of causing disease).
  • the composition comprises a nanoemulsion and an immunogen.
  • the composition comprising a nanoemulsion and an immunogen comprises one or more other compounds or agents including, but not limited to, therapeutic agents, physiologically tolerable liquids, gels, carriers, diluents, adjuvants, excipients, salicylates, steroids, immunosuppressants, immunostimulants, antibodies, cytokines, antibiotics, binders, fillers, preservatives, stabilizing agents, emulsifiers, and/or buffers.
  • An immune response may be an innate (e.g., a non-specific) immune response or a learned (e.g., acquired) immune response (e.g.
  • a composition comprising a nanoemulsion and an immunogen is administered to a subject as a vaccine (e.g., to prevent or attenuate a disease (e.g., by providing to the subject total or partial immunity against the disease or the total or partial attenuation (e.g., suppression) of a sign, symptom or condition of the disease.
  • a vaccine e.g., to prevent or attenuate a disease (e.g., by providing to the subject total or partial immunity against the disease or the total or partial attenuation (e.g., suppression) of a sign, symptom or condition of the disease.
  • adjuvant refers to any substance that can stimulate an immune response (e.g., a mucosal immune response). Some adjuvants cause activation of a cell of the immune system (e.g., an adjuvant can cause an immune cell to produce and secrete a cytokine). Examples of adjuvants that can cause activation of a cell of the immune system include, but are not limited to, the nanoemulsion formulations described herein, saponins purified from the bark of the Q.
  • saponaria tree such as QS21 (a glycolipid that elutes in the 21st peak with HPLC fractionation; Aquila Biopharmaceuticals, Inc., Worcester, Mass.); poly(di(carboxylatophenoxy)phosphazene (PCPP polymer; Virus Research Institute, USA); derivatives of lipopolysaccharides such as monophosphoryl lipid A (MPL; Ribi ImmunoChem Research, Inc., Hamilton, Mont.), muramyl dipeptide (MDP; Ribi) and threonyl-muramyl dipeptide (t-MDP; Ribi); OM-174 (a glucosamine disaccharide related to lipid A; OM Pharma SA, Meyrin, Switzerland); cholera toxin (CT), and Leishmania elongation factor (a purified Leishmania protein; Corixa Corporation, Seattle, Wash.).
  • QS21 a glycolipid that elutes in the 21st peak with HPLC fractionation; Aquila
  • immunogenic compositions described herein are administered with one or more adjuvants (e.g., to skew the immune response towards a Th1 and/or Th2 type response).
  • an amount effective to induce an immune response refers to the dosage level required (e.g., when administered to a subject) to stimulate, generate and/or elicit an immune response in the subject.
  • An effective amount can be administered in one or more administrations (e.g., via the same or different route), applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • the term “under conditions such that said subject generates an immune response” refers to any qualitative or quantitative induction, generation, and/or stimulation of an immune response (e.g., innate or acquired).
  • immune response refers to any detectable response by the immune system of a subject.
  • immune responses include, but are not limited to, an alteration (e.g., increase) in Toll receptor activation, lymphokine (e.g., cytokine (e.g., Th1 or Th2 type cytokines) or chemokine) expression and/or secretion, macrophage activation, dendritic cell activation, T cell (e.g., CD4+ or CD8+T cell) activation, NK cell activation, and/or B cell activation (e.g., antibody generation and/or secretion).
  • lymphokine e.g., cytokine (e.g., Th1 or Th2 type cytokines) or chemokine
  • macrophage activation e.g., dendritic cell activation
  • T cell e.g., CD4+ or CD8+T cell
  • NK cell activation e.g., NK cell activation
  • immune responses include binding of an immunogen (e.g., antigen (e.g., immunogenic polypeptide)) to an MHC molecule and induction of a cytotoxic T lymphocyte (“CTL”) response, induction of a B cell response (e.g., antibody production), and/or T-helper lymphocyte response, and/or a delayed type hypersensitivity (DTH) response (e.g., against the antigen from which an immunogenic polypeptide is derived), expansion (e.g., growth of a population of cells) of cells of the immune system (e.g., T cells, B cells (e.g., of any stage of development (e.g., plasma cells), and increased processing and presentation of antigen by antigen presenting cells.
  • an immunogen e.g., antigen (e.g., immunogenic polypeptide)
  • CTL cytotoxic T lymphocyte
  • B cell response e.g., antibody production
  • T-helper lymphocyte response e.g., T-helper lymphocyte response
  • an immune response may be to immunogens that the subject's immune system recognizes as foreign (e.g., non-self antigens from microorganisms (e.g., pathogens), or self-antigens recognized as foreign).
  • immunogens that the subject's immune system recognizes as foreign
  • immune response refers to any type of immune response, including, but not limited to, innate immune responses (e.g., activation of Toll receptor signaling cascade) cell-mediated immune responses (e.g., responses mediated by T cells (e.g., antigen-specific T cells) and non-specific cells of the immune system) and humoral immune responses (e.g., responses mediated by B cells (e.g., via generation and secretion of antibodies into the plasma, lymph, and/or tissue fluids).
  • innate immune responses e.g., activation of Toll receptor signaling cascade
  • T cells e.g., antigen-specific T cells
  • B cells e.g., via generation and secretion of
  • the term “immunity” refers to protection from disease (e.g., preventing or attenuating (e.g., suppression of) a sign, symptom or condition of the disease) upon exposure to a microorganism (e.g., pathogen) capable of causing the disease.
  • Immunity can be innate (e.g., non-adaptive (e.g., non-acquired) immune responses that exist in the absence of a previous exposure to an antigen) and/or acquired (e.g., immune responses that are mediated by B and T cells following a previous exposure to antigen (e.g., that exhibit increased specificity and reactivity to the antigen)).
  • immunogen and “antigen” are used interchangeably to refer to an agent (e.g., a microorganism (e.g., bacterium, virus or fungus) and/or portion or component thereof (e.g., a protein antigen)) that is capable of eliciting an immune response in a subject.
  • agent e.g., a microorganism (e.g., bacterium, virus or fungus) and/or portion or component thereof (e.g., a protein antigen)
  • immunogens elicit immunity against the immunogen (e.g., microorganism (e.g., pathogen or a pathogen product)) when administered in combination with a nanoemulsion of the present invention.
  • Burkholderia antigen refers to a component or product of a bacteria of the genus Burkholderia that elicits an immune response when administered to a subject.
  • An antigen may be a component or product derived from an organism (e.g., bacteria of the genus Burkholderia ) including, but not limited to, polypeptides, peptides, proteins, nucleic acids, membrane fractions, and polysaccharides.
  • the term “enhanced immunity” refers to an increase in the level of adaptive and/or acquired immunity in a subject to a given immunogen (e.g., microorganism (e.g., pathogen)) following administration of a composition relative to the level of adaptive and/or acquired immunity in a subject that has not been administered the composition.
  • a given immunogen e.g., microorganism (e.g., pathogen)
  • the terms “purified” or “to purify” refer to the removal of contaminants or undesired compounds from a sample or composition.
  • the term “substantially purified” refers to the removal of from about 70 to 90%, up to 100%, of the contaminants or undesired compounds from a sample or composition.
  • administering refers to the act of giving a drug, prodrug, or other agent, or therapeutic treatment (e.g., a composition of the present invention) to a physiological system (e.g., a subject or in vivo, in vitro, or ex vivo cells, tissues, and organs).
  • a physiological system e.g., a subject or in vivo, in vitro, or ex vivo cells, tissues, and organs.
  • co-administration refers to the administration of at least two agent(s) (e.g., a nanoemulsion and one or more other pharmaceutically acceptable substances (e.g., a second nanoemulsion)) or therapies to a subject.
  • the co-administration of two or more agents or therapies is concurrent.
  • a first agent/therapy is administered prior to a second agent/therapy.
  • co-administration can be via the same or different route of administration.
  • formulations and/or routes of administration of the various agents or therapies used may vary. The appropriate dosage for co-administration can be readily determined by one skilled in the art.
  • co-administration when agents or therapies are co-administered, the respective agents or therapies are administered at lower dosages than appropriate for their administration alone.
  • co-administration is especially desirable in embodiments where the co-administration of the agents or therapies lowers the requisite dosage of a potentially harmful (e.g., toxic) agent(s), and/or when co-administration of two or more agents results in sensitization of a subject to beneficial effects of one of the agents via co-administration of the other agent.
  • co-administration is preferable to treat and/or prevent infection by more than one type of infectious agent (e.g., bacteria and/or viruses).
  • topically refers to application of a compositions of the present invention (e.g., a composition comprising a nanoemulsion) to the surface of the skin and/or mucosal cells and tissues (e.g., alveolar, buccal, lingual, masticatory, vaginal or nasal mucosa, and other tissues and cells which line hollow organs or body cavities).
  • a compositions of the present invention e.g., a composition comprising a nanoemulsion
  • mucosal cells and tissues e.g., alveolar, buccal, lingual, masticatory, vaginal or nasal mucosa, and other tissues and cells which line hollow organs or body cavities.
  • compositions described herein can be applied using any pharmaceutically acceptable method, such as for example, intranasal, buccal, sublingual, oral, rectal, ocular, parenteral (intravenously, intradermally, intramuscularly, subcutaneously, intracisternally, intraperitoneally), pulmonary, intravaginal, locally administered, topically administered, mucosally administered, via an aerosol, or via a buccal or nasal spray formulation.
  • the nanoemulsion vaccines described herein can be formulated into any pharmaceutically acceptable dosage form, such as a liquid dispersion, gel, aerosol, pulmonary aerosol, nasal aerosol, ointment, cream, semi-solid dosage form, and a suspension.
  • the composition may be a controlled release formulation, sustained release formulation, immediate release formulation, or any combination thereof.
  • compositions that do not substantially produce adverse allergic or immunological reactions when administered to a host (e.g., an animal or a human). Such formulations include dips, sprays, seed dressings, stem injections, sprays, and mists.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, wetting agents (e.g., sodium lauryl sulfate), isotonic and absorption delaying agents, disintegrants (e.g., potato starch or sodium starch glycolate), and the like.
  • the term “pharmaceutically acceptable salt” refers to any salt (e.g., obtained by reaction with an acid or a base) of a composition of the present invention that is physiologically tolerated in the target subject. “Salts” of the compositions of the present invention may be derived from inorganic or organic acids and bases.
  • acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, sulfonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like.
  • Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compositions of the invention and their pharmaceutically acceptable acid addition salts.
  • bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW 4 + , wherein W is C 1-4 alkyl, and the like.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • W is C 1-4 alkyl
  • salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosy
  • salts include anions of the compounds of the present invention compounded with a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a C 1-4 alkyl group), and the like.
  • a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a C 1-4 alkyl group), and the like.
  • salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable.
  • salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
  • salts of the compositions of the present invention are contemplated as being pharmaceutically acceptable.
  • salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable composition.
  • the terms “at risk for disease” and “at risk for infection” refer to a subject that is predisposed to experiencing a particular disease and/or infection. This predisposition may be genetic (e.g., a particular genetic tendency to experience the disease, such as heritable disorders), or due to other factors (e.g., environmental conditions, exposures to detrimental compounds present in the environment, etc.). Thus, it is not intended that the present invention be limited to any particular risk (e.g., a subject may be “at risk for disease” simply by being exposed to and interacting with other people that carry a risk of transmitting a pathogen), nor is it intended that the present invention be limited to any particular disease and/or infection.
  • nasal application means applied through the nose into the nasal or sinus passages or both.
  • the application may, for example, be done by drops, sprays, mists, coatings or mixtures thereof applied to the nasal and sinus passages.
  • “Pulmonary application” and “pulmonary administration” refers to any means of applying a composition of the present invention to the pulmonary system of a subjet.
  • the present invention is not limited to any particular means of administration. Indeed, a variety of means are contemplated to be useful for pulmonary administration including those described herein.
  • kits refers to any delivery system for delivering materials.
  • such delivery systems include systems that allow for the storage, transport, or delivery of the compositions and/or supporting materials (e.g., written instructions for using the materials, etc.) from one location to another.
  • kits include one or more enclosures (e.g., boxes) containing the relevant nanoemulsions and/or supporting materials.
  • fragment kit refers to delivery systems comprising two or more separate containers that each contain a subportion of the total kit components. The containers may be delivered to the intended recipient together or separately.
  • a first container may contain a composition comprising a nanoemulsion for a particular use, while a second container contains a second agent (e.g., an antibiotic or spray applicator).
  • a second agent e.g., an antibiotic or spray applicator
  • any delivery system comprising two or more separate containers that each contains a subportion of the total kit components are included in the term “fragmented kit.”
  • a “combined kit” refers to a delivery system containing all of the components of a composition needed for a particular use in a single container (e.g., in a single box housing each of the desired components).
  • kit includes both fragmented and combined kits.
  • pathogenic microorganisms initiate infection by attaching to mucosal epithelial cells lining the gastro-intestinal, oropharyngeal, respiratory or genito-urinacy tracts.
  • Some pathogens such as influenza virus, Bordetella pertussis , or Vibrio cholerae , remain at or within the mucosal tissue, while others, such as Salmonella typhi or hepatitis A virus, possess mechanisms permitting penetration into deeper tissues and spread systemically.
  • Specific and non-specific defense mechanisms of the mucous membranes provide first line protection against both types of pathogen.
  • Non-specific effectors include resident macrophages, antimicrobial peptides, lactoferrin and lysozyme, extremes of pH, bile acids, digestive enzymes, mucus, shedding of epithelial cells, flushing mechanisms (peristalsis, ciliary beating, micturation, etc.) and competition from local flora.
  • successful pathogens have generally evolved means to survive the non-specific defenses present at the site they infect and it is the secretory immune system which plays a major role in protecting against diseases caused by a number of bacterial and viral pathogens, and is probably a major effector against pathogens that are restricted to mucosal surfaces. For organisms that spread systemically, both local and systemic immune responses are desirable for optimum immunity.
  • certain microbes e.g., bacteria
  • CF Cystic fibrosis
  • asthma a normal and/or healthy immune system does not function properly for one reason or another.
  • chemotherapeutic therapy a host of other conditions lead to malfunctioning and/or attenuation of immune responses that would normally function to protect against and clear microbes capable of causing pathology in a healthy subject.
  • Pseudomonas aeruginosa is an opportunistic pathogen that infects the immunocompromised, elderly, cancer chemotherapy patients, and individual suffering from CF.
  • P. aeruginosa is trapped in thickened, dehydrated, hypoxic mucus lining in airway epithelia.
  • Morphologic data suggests that the airway lumen of CF patients harbor P. aeruginosa biofilms that are characterized as spherical microcolonies.
  • B. pertussis differs from B. bronchiseptica and B. parapertussis in the nature of the toxins it produces, B. bronchiseptica and B. parapertussis do produce active toxins (See Hausman, S. Z. et al., 1996, Infect. Immun. 64: 4020-4026), and there is some evidence to indicate that B. pertussis organisms can covert to the B. parapertussis phenotype (Gilchrist, M. J. R., 1991, “ Bordetella ”, in Manual of Clinical Microbiology, 5th ed., Balows, A. et al., eds., American Society for Microbiology, Washington, D.C.).
  • compositions and methods for the treatment of respiratory infections may be used to treat and/or prevent respiratory infection (e.g., in cystic fibrosis patients) caused by one or more of pseudomonas (e.g., P. aeruginosa, P. paucimobilis, P. putida, P. fluorescens , and P.
  • pseudomonas e.g., P. aeruginosa, P. paucimobilis, P. putida, P. fluorescens , and P.
  • compositions and methods of the present invention find use in the treatment and/or prevention of a host of respiratory infections (e.g., respiratory infections of the upper respiratory tract (e.g., nose, ears, sinuses, and throat) and the lower respiratory tract (e.g., trachea, bronchial tubes, and lungs)).
  • respiratory infections e.g., respiratory infections of the upper respiratory tract (e.g., nose, ears, sinuses, and throat) and the lower respiratory tract (e.g., trachea, bronchial tubes, and lungs)
  • microbes e.g., killed and/or attenuated in growth (e.g., within the respiratory tract of a subject) are provided below.
  • the present invention provides a composition comprising a nanoemulsion, and methods of pulmonary administration of the same to prevent and/or treat respiratory infection.
  • the nanoemulsion comprises ethylenediaminetetraacetic acid (EDTA).
  • EDTA ethylenediaminetetraacetic acid
  • the present invention is not limited by the amount of EDTA utilized. In some embodiments, 0.01-0.1 mM, 0.1-1.0 mM, 1.0-1.5 mM, 1.5-2.5 mM, 2.5-5.0 mM, 5.0-10.0 mM, 10-20 mM, 20-30 mM, or 30-50 mM EDTA is used. However, the present invention is not limited to this amount of EDTA.
  • the composition is co-administered with a hypertonic salt (e.g., sodium chloride) solution (e.g., a 6-7%, 1-3%, 3-6%, 0.1-1%, or more than 7% salt solution (e.g., NaCl solution)).
  • a hypertonic salt e.g., sodium chloride
  • the composition comprises a 20% nanoemulsion solution.
  • the composition comprises greater than 20% (e.g., 25%, 30%, or more) nanoemulsion solution.
  • the composition comprises less than 20% (e.g., 15%, 10% or less) nanoemulsion solution.
  • a composition comprises less than 10% nanoemulsion. In some embodiments, a composition comprises more than 20% nanoemulsion. In some embodiments, the composition comprises 10 mM EDTA. In some embodiments, the composition comprises 20 mM EDTA. In some embodiments, a composition of the present invention comprises any of the nanoemulsions described herein. In some embodiments, a composition comprising a nanoemulsion utilized to treat bacteria (e.g., present in pulmonary space of a subject (e.g., biofilm forming bacteria)) comprises P 407 5EC. In some embodiments, a composition comprising a nanoemulsion comprises W 80 5EC.
  • nanoemulsion alone or in combination with EDTA (e.g., 10-20 mM EDTA) was able to achieve complete killing of 10 6 bacteria in PBS in 60 minutes (See Examples 2-4).
  • hypertonic saline e.g., 6-7% NaCl
  • the killing ability of the nanoemulsion was surprisingly and strikingly enhanced, achieving complete killing within 15 minutes while in the presence of 20 mM EDTA (See Example 2).
  • nanoemulsions comprising a lower concentration of EDTA were able to achieve complete killing of bacteria in 30 minutes in the presence of hypertonic saline.
  • the present invention provides that a nanoemulsion composition can be used to kill (e.g., completely) bacteria over a short time period (e.g., less than 60 minutes, less than 30 minutes, or less than 15 minutes).
  • a short time period e.g., less than 60 minutes, less than 30 minutes, or less than 15 minutes.
  • compositions of the present invention are able to eradicate a mixed population of bacteria.
  • toxicity studies performed during the development of embodiments of the present invention characterized the nanoemulsion compositions as being safe and causing no detectable harm to a subject (e.g., no histological changes and/or detectable pathology (See, e.g., Example 1)).
  • compositions comprising nanoemulsions of the present invention are able to treat (e.g., kill and/or inhibit growth of) bacterial species that are generally not virulent in healthy persons, but that are opportunists that cause severe and chronic respiratory tract infections (e.g., in individuals with cystic fibrosis (CF)). Unremitting infection with these species results in inflammation and progressive lung disease that culminates in pulmonary failure, the leading cause of death for CF patients.
  • Effective therapy of pulmonary infection in CF has been severely limited by the broad spectrum antimicrobial resistance exhibited by these species, which are among the most drug-resistant bacteria encountered in human infection. The site of infection in CF presents another important obstacle to effective therapy.
  • Infecting bacteria primarily reside within the airway lumen in sputum, airway epithelial surface fluid, and the bronchial mucosa (ref). The penetration of systemically delivered antimicrobials to this infected site is generally poor. Treatment is further hampered by bacterial biofilm formation, which is believed to occur in the airways of infected patients, and by the exceptionally viscous secretions that characterize the CF respiratory tract.
  • a mechanism of bacterial and/or viral killing utilizing compositions comprising nanoemulsions of the present invention involves fusion of the emulsion with microorganism lipid membranes, leading to rapid osmotic disruption and cell lysis (See, e.g., Hamouda and Baker, J Appl Microbiol. 2000 September; 89(3):397-403).
  • electrostatic attraction e.g., provided by cationic surface charge of CPC
  • electrostatic attraction overcomes the LPS-mediated resistance of gram-negative bacteria to neutral and anionic detergents (See, e.g., Hamouda and Baker, J Appl Microbiol. 2000 September; 89(3):397-403).
  • bactericidal activity of a composition comprising a nanoemulsion e.g., P 407 5EC (e.g., against gram-negative and/or gram-positive bacteria) is enhanced by the addition of EDTA.
  • EDTA chelates divalent cations that stabilize outer membrane LPS thereby facilitating interactions with the cationic emulsion, an interaction that leads to membrane permeabilization and lysis as well as augmenting transmembrane diffusion of macromolecules (See, e.g., Rabinovich-Guilatt et al., J Drug Target. 2004; 12(9-10):623-33, Vaara, Microbiol Rev. 1992 September; 56(3):395-411).
  • the present invention provides compositions comprising P 407 5EC and hypertonic saline, and methods of using the same (e.g., via inhalation (e.g., post nebulizing the solution)) for treating one or more types of pulmonary bacterial infections (e.g., to treat bacteria and/or bacterial biofilms (e.g., that possess resistance to conventional treatments) without increasing the already high treatment burden for CF patients).
  • inhalation e.g., post nebulizing the solution
  • pulmonary bacterial infections e.g., to treat bacteria and/or bacterial biofilms (e.g., that possess resistance to conventional treatments) without increasing the already high treatment burden for CF patients.
  • compositions comprising nanoemulsions e.g., P 407 5EC and saline solution
  • Bcc Burkholderia cepacia complex
  • Infection with Burkholderia cepacia species is particularly refractory to antimicrobial therapy and associated with increased rates of morbidity and mortality in CF.
  • Infection with Bcc is also regarded by many CF care centers as an absolute contraindication to lung transplantation.
  • compositions comprising nanoemulsions were effective to treat (e.g., kill and/or inhibit growth of) B. multivorans and B. cenocepacia which account for the majority of Bcc infection in CF (See, e.g., Reik J Clin Microbiol. 2005 June; 43(6):2926-8).
  • Compositions comprising nanoemulsions were effective to treat (e.g., kill and/or inhibit growth of) B. gladioli , which although not a member of the Bcc, is being recovered with increasing frequency from CF patients.
  • compositions comprising nanoemulsions were effective to treat (e.g., kill and/or inhibit growth of) Acinetobacter , which although currently infrequently recovered in CF, appears to be an emerging pathogen in this patient population.
  • Acinetobacter which although currently infrequently recovered in CF, appears to be an emerging pathogen in this patient population.
  • the great majority (94%) of isolates tested in Example 3 were recovered from cultures of respiratory specimens from persons with CF.
  • the panel also included one representative isolate from each of five previously described so-called epidemic lineages, each of which has been identified as infecting multiple CF patients. These included the B.
  • cenocepacia ET12 Johnson et al., J Clin Microbiol 1994; 32:924-30.
  • PHDC Choen et al., J Pediatr 2001; 139:643-9
  • Midwest Coenye and LiPuma, J. Infect. Dis. 185:1454-1462 lineages, as well as the B. multivorans OHBM lineage (Biddick et al., FEMS Microbiol Lett 2003; 228:57-62) and the B. dolosa SLC6 lineage (Biddick et al., FEMS Microbiol Lett 2003; 228:57-62).
  • compositions comprising nanoemulsions e.g., P 407 5EC and saline solution
  • nanoemulsions e.g., P 407 5EC and saline solution
  • compositions comprising nanoemulsions were effective to treat (e.g., kill and/or inhibit growth of) strains that were found in previous susceptibility testing to be multi-drug resistant (e.g., defined as resistant to all antibiotics tested in two of three antibiotic classes: lactams (including carbapenems), aminoglycosides, and quinolones) as well as panresistant strains (See Example 3).
  • Genotyping analyses of all isolates was performed to confirm that each sample treated was a distinct strain.
  • P 407 5EC showed very good activity against the strains tested.
  • all strains in the test panel were inhibited by a P 407 5EC concentration of ⁇ 125 ⁇ g/ml, or a 1:16 dilution of the starting material; 59% of strains were inhibited by a concentration of ⁇ 31.2 ⁇ g/ml, a 1:64 dilution of P 407 5EC.
  • Multi-drug resistant or panresistant strains did not show decreased susceptibility to P 407 5EC, demonstrating MIC 90 s of 125 ⁇ g/ml and 62.5 ⁇ g/ml, respectively.
  • Burkholderia species tended to be slightly less susceptible to P 407 5EC than the other species examined. Sixteen of the 18 strains requiring the highest MICs were Burkholderia . Conversely, 33 of the 38 strains with the lowest tested MIC (15.6 ⁇ g/ml) were non- Burkholderia species. No striking differences in P 407 5EC activity was observed among the 10 Burkholderia species examined, although the two strains requiring the highest MICs (250 ⁇ g/ml and 500 ⁇ g/ml) were both B. cenocepacia .
  • a composition comprising a nanoemulsion (e.g., P 407 5EC) of the present invention can be utilized individually, or in combination with other antimicrobials, to kill and/or inhibit growth of bacteria.
  • CF sputum like biofilm, is reported to antagonize the activity of antibacterial drugs (See, e.g., Hunt et al., Antimicrob Agents Chemother. 1995 January; 39(1):34-9).
  • Glycoproteins such as mucin, which compose 2-3% of the dry weight of sputum, and high molecular weight DNA are present in elevated levels resulting in exceptionally viscous sputum that provides a physical barrier protecting bacteria.
  • these macromolecules bind and sequester antibiotics while small cationic molecules and the decreased pH of CF sputum block drug penetration into bacteria and reduce drug bioactivity.
  • the strategy of increasing drug dosing to overcome these obstacles is limited by drug toxicity.
  • the activity of P 407 5EC against bacteria suspended in media containing 43% sputum was decreased with bactericidal concentrations 2- to 32-fold greater than the respective planktonic MBCs without sputum.
  • the sputum-MBCs were identical to (6 of 12) or within one dilution of (6 of 12) the MBECs obtained with biofilm grown bacteria.
  • the activity of P 407 5EC was similarly antagonized by both CF sputum and biofilm growth, it remained bactericidal for all the strains tested under both test conditions (See FIG. 10 ).
  • the present invention provides compositions comprising nanoemulsions (e.g., P 407 5EC) and methods of using the same for antimicrobial treatment for infection due to CF-related opportunistic pathogens.
  • nanoemulsion compositions of the present invention are rapidly bactericidal, and active against bacteria whether grown planktonically or as a biofilm, or in the presence of CF sputum.
  • compositions comprising nanoemulsions of the present invention are exceptionally stable, unchanged after nebulization, and broadly microbicidal.
  • the development of resistance to a nanoemulsion composition of the present invention has not been observed by any bacterial species examined to date.
  • the present invention provides that P 407 5EC can be used effectively as an inhaled antimicrobial.
  • compositions comprising a nanoemulsion of the present invention can be utilized as a therapeutic and/or antimicrobial agent (e.g., to kill and/or inhibit growth of) bacterial biofilms in any clinical and/or industrial setting.
  • biofilms Multiple species of bacteria exist that are able to form biofilms. For example, bacteria that adhere to implanted medical devices or damaged tissue often encase themselves in a hydrated matrix of polysaccharide and protein to form biofilm. Biofilms pose a serious problem for public health because of the increased resistance of biofilm-associated organisms to antimicrobial agents and the association of infections with these organisms in patients with indwelling medical devices or damaged tissue. Antibiotic resistance of bacteria growing in biofilms contributes to the persistence and chronic nature of infections such as those associated with implanted medical devices. The mechanisms of resistance in biofilms are different from the now familiar plasmids, transposons, and mutations that confer innate resistance to individual bacterial cells. In biofilms, resistance seems to depend on multicellular strategies.
  • Biofilms are complex communities of microorganisms attached to surfaces or associated with interfaces or damaged tissue.
  • planktonic (free-swimming) bacteria it is now widely recognized that most bacteria found in natural, clinical, and industrial settings persist in association with surfaces as biofilms.
  • these microbial communities are often composed of multiple species that interact with each other and their environment.
  • the determination of biofilm architecture particularly the spatial arrangement of microcolonies (clusters of cells) relative to one another, has profound implications for the function of these complex communities.
  • the biofilm matrix is a dynamic environment in which the component microbial cells appear to reach homeostasis and are optimally organized to make use of all available nutrients.
  • the matrix therefore shows great microheterogeneity, within which numerous microenvironments can exist.
  • Biofilm formation is believed to be a two-step process in which the attachment of bacterial cells to a surface is followed by growth dependent accumulation of bacteria in multilayered cell clusters.
  • exopolysaccharides provide the matrix framework, a wide range of enzyme activities can be found within the biofilm, some of which greatly affect structural integrity and stability.
  • the fibrinogen and fibronectin of host plasma cover the surface of a medical implant or damaged tissue and are identified by constitutively expressed microbial surface components, which mediate the initial attachment of bacteria to the surface of the biomaterial or damaged tissue.
  • a specific gene locus in the bacteria cells called the intracellular adhesion (ica) locus, activates the adhesion of bacteria cells to each other, forming the secondary layers of the biofilm.
  • the ica locus is responsible for the expression of the capsular polysaccharide operon, which in turn activates polysaccharide intercellular adhesion (PIA), via the sugar poly-N-succinylglucosamine (PNSG), a-1,6-linked glucosaminoglycan.
  • PIA polysaccharide intercellular adhesion
  • PNSG sugar poly-N-succinylglucosamine
  • a-1,6-linked glucosaminoglycan a-1,6-linked glucosaminoglycan.
  • Staphylococcus aureus is a highly virulent human pathogen. Both S. aureus and coagulase-negative staphylococci have emerged as major nosocomial pathogens associated with biofilm formation on implanted medical devices and damaged tissue. These organisms are among the normal carriage flora of human skin and mucous membranes, making them prevalent complications during and after invasive surgery or prolonged hospital stays. As bacteria carried on both healthy and sick people, staphylococci are considered opportunistic pathogens that invade patients via open wounds and via biomaterial implants.
  • Biofilm infections associated with S. aureus are a significant cause of morbidity and mortality, particularly in settings such as hospitals, nursing homes and infirmaries. Patients at risk include infants, the elderly, the immuno-compromised, the immuno-suppressed, and those with chronic conditions requiring frequent hospital stays. Patients with intravascular and other implanted prosthetic devices are at even greater risk from staphylococcal infections because of compromised immune systems and the introduction of foreign bodies, which serve to damage tissue and/or act as a surface for the formation of biofilms. Such infections can have chronic, if not fatal, implications.
  • the causes of biofilm resistance to antibiotics include the failure of some antimicrobial agents to penetrate all the layers of a biofilm, the slow-growth rate of certain biofilm cells that make them less susceptible to antimicrobial agents requiring active bacterial growth, and the expression of gene patterns by the bacterial cells embedded in the biofilm that differ from the genes expressed in their planktonic (free-swimming) state. These differences in biofilm-associated bacteria render antimicrobial agents that work effectively to kill planktonic bacteria ineffective in killing biofilm-associated bacteria. Often the only way to treat biofilms (e.g., associated with catheters or prosthetic devices) is the removal of the contaminated device, which may require additional surgery and present further risks to patients.
  • biofilms refer to an aggregate of microorganisms with an extracellular matrix that facilitates adhesion to, and colonization and growth of the aggregate on a surface, such as an internal or external tissue or organ.
  • Biofilms can be comprised of bacteria, fungi, yeast, protozoa, or other microorganisms.
  • Bacterial biofilms typically display high resistance to antibiotics, often up to 1,000-times greater resistance than the same bacteria not growing in a biofilm.
  • compositions and methods of the invention are utilized to treat (e.g., kill and/or inhibit growth of) and/or prevent biofilms on and/or within a subject (e.g., within the pulmonary system, on internal organs or tissue (e.g., the bladder, kidney, heart, middle ear, sinuses, a joint, the eye), on an external tissue (e.g., the skin), and/or oral surfaces such as teeth, tongue, oral mucosa, or gums.
  • a subject e.g., within the pulmonary system, on internal organs or tissue (e.g., the bladder, kidney, heart, middle ear, sinuses, a joint, the eye), on an external tissue (e.g., the skin), and/or oral surfaces such as teeth, tongue, oral mucosa, or gums.
  • compositions and methods of the invention may be used to treat a biofilm-associated condition such as a soft-tissue infection, chronic sinusitis, endocarditis, osteomyelitis, urinary tract infection, chronic bacterial vaginosis, dental plaque or halitosis, infection of prosthetic device and/or catheter, bacterial keratitis, or prostatitis.
  • a biofilm-associated condition such as a soft-tissue infection, chronic sinusitis, endocarditis, osteomyelitis, urinary tract infection, chronic bacterial vaginosis, dental plaque or halitosis, infection of prosthetic device and/or catheter, bacterial keratitis, or prostatitis.
  • compositions of the present invention can be utilized to treat (e.g., kill and/or inhibit growth of) any one or more Gram-positive and Gram-negative bacterial species.
  • compositions and methods of the present invention can be utilized to kill and/or inhibit growth of a number of bacterial species including, but not limited to, Staphylococcus aureus , coagulase negative staphylococci such as Staphylococcus epidermis, Streptococcus pyogenes (Group A), Streptococcus species (viridans group), Streptococcus agalactiae (group B), S.
  • Streptococcus anaerobic species
  • Streptococcus pneumoniae Enterococcus species
  • Bacillus anthracis Corynebacterium diptheriae
  • Corynebacterium species which are diptheroids diptheroids (aerobic and anaerobic)
  • Listeria monocytogenes Clostridium tetani
  • Clostridium difficile Escherichia coli
  • Enterobacter species Proteus mirablis
  • Pseudomonas aeruginosa Klebsiella pneumoniae, Salmonella, Shigella, Serratia, Campylobacter jejuni, Neisseria, Branhamella catarrhalis , and Pasteurella.
  • compositions and methods of the present invention can be utilized to treat (e.g., kill and/or inhibit growth of) organisms capable of forming biofilms including, but not limited to, dermatophytes (e.g, Microsporum species such as Microsporum canis, Trichophyton species such as Trichophyton rubrum and Trichophyton mentagrophytes ), yeasts (e.g., Candida albicans, Candidaparapsilosis, Candida glabrata, Candida tropicalis , and other Candida species including drug resistant Candida species), Epidermophytonfloccosum, Malasseziafuurfur ( Pityropsporon orbiculare, Pityropsporon ovale ) Cryptococcus neoformans, Aspergillusfumigatus and other Aspergillus species, Zygomycetes ( Rizopus, Mucor ), hyalohyphomycosis ( Fusarium species), Paracoccidiodes brasiliensis, Blastmyces dermatitides, His
  • the present invention provides a method for treating a subject possessing a biofilm (e.g., possessing an indwelling prosthetic device or catheter, wherein the indwelling prosthetic device or catheter is in contact with a biofilm, or wherein the subject has an infection (e.g., respiratory infection) within which a biofilm resides) comprising administering to the subject a composition comprising a nanoemulsion (e.g., P 407 5EC) under conditions such that the biofilm is altered and/or bacteria residing within the biofilm are killed and/or their growth is inhibited.
  • altering the biofilm comprises eradicating the biofilm.
  • altering the biofilm comprises killing bacteria involved in forming the biofilm.
  • the bacteria comprise S. aureus, S. epidermidis , antibiotic resistant bacteria (e.g., methicillin resistant, vancomycin resistant, etc.), and/or other type of bacteria described herein.
  • the composition comprising a nanoemulsion e.g., P 407 5EC
  • the antibacterial agents are selected from the group comprising, but not limited to, antibiotics, antibodies, antibacterial enzymes, peptides, and lanthione-containing molecules.
  • the antibiotic interferes with or inhibits cell wall synthesis.
  • the antibiotic is selected from the group including, but not limited to, ⁇ -lactams, cephalosporins, glycopeptides, aminoglycosides, sulfonomides, macrolides, folates, polypeptides and combinations thereof.
  • the antibiotic interferes with protein synthesis (e.g., glycosides, tetracyclines and streptogramins).
  • the present invention is not limited by the number of doses of composition comprising nanoemulsion administered. In some embodiments, multiple doses are administered on separate days. In some embodiments, the multiple doses are administered on the same day. In some embodiments, a composition comprising a nanoemulsion described herein is administered continuously.
  • co-administration with a composition comprising a nanoemulsion permits administering a lower dose of an antibacterial agent than would be administered without co-administration of a composition comprising a nanoemulsion.
  • the composition comprising a nanoemulsion described herein is administered using a nebulizer.
  • administration is intramuscularly, subcutaneously, locally, directly into an infected site, directly onto an indwelling prosthetic device (e.g., a shunt, stent, scaffold for tissue construction, feeding tube, punctual plug, artificial joint, pacemaker, artificial valve, etc.) or catheter.
  • administration is directly through a catheter.
  • the present invention is not limited by the type of microbe treated. Indeed a variety of microbial pathogens can be treated (e.g, killed (e.g., completely killed)) and/or the growth thereof prevented and/or attenuated in a subject using the compositions and methods of the present invention including, but not limited to, bacteria, viruses, and fungi described herein.
  • microbial pathogens e.g, killed (e.g., completely killed)
  • the growth thereof prevented and/or attenuated in a subject using the compositions and methods of the present invention including, but not limited to, bacteria, viruses, and fungi described herein.
  • the present invention also provides compositions and methods for treating (e.g., killing and/or inhibiting growth of) organisms that heretofore display resistance to a broad spectrum of antibiotics (e.g., species of the genus Acinetobacter ).
  • antibiotics e.g., species of the genus Acinetobacter
  • Acinetobacter species are generally considered nonpathogenic to healthy individuals. However, several species persist in hospital environments and cause severe, life-threatening infections in compromised patients (See, e.g., Gerischer U (editor). (2008). Acinetobacter Molecular Biology, 1st ed., Caister Academic Press). The spectrum of antibiotic resistances of these organisms together with their survival capabilities make them a threat to hospitals as documented by recurring outbreaks both in highly developed countries and elsewhere. Infections occur in immunocompromised individuals, and the strain A. baumannii is the second most commonly isolated nonfermenting bacteria in human specimens. Acinetobacter is frequently isolated in nosocomial infections and is especially prevalent in intensive care units, where both sporadic cases as well as epidemic and endemic occurrence is common. A.
  • baumannii is a frequent cause of nosocomial pneumonia, especially of late-onset ventilator associated pneumonia. It can cause various other infections including skin and wound infections, bacteremia, and meningitis. A. lwoffi is also causative of meningitis. A. baumannii can survive on the human skin or dry surfaces for weeks.
  • Acinetobacter species are innately resistant to many classes of antibiotics, including penicillin, chloramphenicol, and often aminoglycosides. Resistance to fluoroquinolones has been reported during therapy and this has also resulted in increased resistance to other drug classes mediated through active drug efflux. A dramatic increase in antibiotic resistance in Acinetobacter strains has been reported by the CDC and the carbapenems are recognized as the gold-standard and/or treatment of last resort. An increase in resistance to the carbapenems leaves very little treatment option although there has been some success reported with polymyxin B. Acinetobacter species are unusual in that they are sensitive to sulbactam; sulbactam is most commonly used to inhibit bacterial beta-lactamase, but this is an example of the antibacterial property of sulbactam itself.
  • compositions and methods of the present invention are utilized to treat (e.g., kill and/or inhibit growth of) bacteria of the Acinetobacter species (e.g., individually or in combination with other treatments (e.g., carbapenems, polymyxin B, and/or sulbactam)).
  • bacteria of the Acinetobacter species e.g., individually or in combination with other treatments (e.g., carbapenems, polymyxin B, and/or sulbactam)).
  • Cystic fibrosis is a life-threatening disorder that causes severe lung damage due to a defective transmembrane protein called CFTR responsible for the balance of electrolytes. Thick mucus forms plugging the tubes, ducts and passageways in the lungs. This environment is ideal for opportunistic bacteria to establish biofilm communities, leading to respiratory infections. Systemically-administered antibiotics can decrease the frequency and severity of exacerbations; however, the bacteria are never be completely eradicated from the airways and the lungs. Nebulized antibiotics are used, but resistance emergence and/or colonization of different resistant species is a major concern.
  • Cystic fibrosis results in the functional impairment of innate respiratory defense mechanisms, providing an environment for colonization of pathogenic bacterial species such as Staphylococcus aureus and Haemophilus influenzae , and a number of opportunistic species such as Pseudomonas aeruginosa, Achromobacter xylosoxidans, Stenotrophomonas maltophilia, Ralstonia spp., Pandoraea spp., and the Burkholderia cepacia complex (Bcc) species (See, e.g., LiPuma et al., (2009) Antimicrob Agents Chemother 53, 249-255).
  • pathogenic bacterial species such as Staphylococcus aureus and Haemophilus influenzae
  • opportunistic species such as Pseudomonas aeruginosa, Achromobacter xylosoxidans, Stenotrophomonas maltophilia,
  • the Bcc comprises a group of at least 17 phylogenetically related saprophytic gram-negative bacilli, most of which can form biofilm (See, e.g., Al Bakri et al., 2004, Journal of Applied Microbiology 96, 455-463; Eberl and Tummler, 2004, International Journal of Medical Microbiology 294, 123-131; LiPuma et al., (2009) Antimicrob Agents Chemother 53, 249-255; and Tomlin et al., 2004, Journal of Microbiological Methods 57, 95-106). They are particularly difficult to treat and are associated with increased rates of morbidity and mortality in CF patients.
  • the present invention is directed to a novel broad-spectrum antimicrobial nanoemulsion (NE) and uses thereof.
  • the NE kills pathogens by interacting with their membranes. This physical kill-on-contact mechanism significantly reduces any concerns about resistance.
  • the NE is formulated from pharmaceutically approved safe ingredients.
  • Example 8 the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of a against four genera of problematic bacteria in CF patients was evaluated: Pseudomonas, Burkholderia, Acinetobacter and Stenotrophomonas .
  • Example 8 also describes evaluating potential synergy between the NE and other antimicrobials.
  • P. aeruginosa, B. cenocepacia, A. baumannii and S. maltophilia which were evaluated in Example 8, are important respiratory pathogens implicated in acute exacerbations of cystic fibrosis patients.
  • the activity of a nanoemulsion is defined herein in terms of its minimum inhibitory concentration (MIC), and/or minimum bactericidal concentration (MBC), both in comparison to traditionally used antimicrobial medicines
  • Example 8 show that the MIC 90 /MBC 90 values for the NE tested were 8/64 ⁇ g/ml for P. aeruginosa, 64/>514 ⁇ g/ml for B. cenocepacia, 8/64 ⁇ g/ml for A. baumannii and 8/32 ⁇ g/ml for S. maltophilia .
  • Colistin had MIC 90 /MBC 90 values of 2/8, >32/>32, 1/>16 and >32/>32 for P. aeruginosa, B. cenocepacia, A. baumannii and S. maltophilia , respectively.
  • Cefepime, imipenem, levofloxacin and tobramycin had MIC 90 /MBC 90 values of ⁇ 32/ ⁇ 32, ⁇ 32/>32, 16/16 and >32/>32 ⁇ g/ml, respectively, against all strains. These results are significant as in contrast to conventional drugs such as colistin, cefepime, imipenem, levofloxacin and tobramycin used to treat CF and which can exhibit significant side effects and potential toxicity with increasing dosage, NE are completely non-toxic.
  • Example 8 Also evaluated in Example 8 below was synergy data. Specifically, also evaluated was the fractional inhibitory concentration (FIC) index when in combination with another antimicrobial and the fractional bactericidal concentration (FBC) index when in combination with another antimicrobial.
  • FIC fractional inhibitory concentration
  • FBC fractional bactericidal concentration
  • the FIC and the FBC are calculated to make the judgment on synergy, antagonism or indifference of the nanoemulsion in combination. For this determination, ten strains of Burkholderia, Stenotrophomonas and 10 strains of Acinetobacter were tested to determine a shift in MIC when P 407 5EC+EDTA was in combination with either colistin or tobramycin, two traditional antimicrobials used in the lungs of CF patients to treat chronic lung infections.
  • FIC fractional inhibitory concentration
  • FBC fractional bactericidal concentration
  • P 407 5EC+EDTA in combination with colistin was found to be indifferent, only 30% synergy in by the FIC and 10% by the FBC, but when in combination with tobramycin, 50% synergy in by the FIC and 20% by the FBC.
  • Example 8 generated a short time-kill curve to produce samples to be used in pictures using electron microscopy for a strain of Burkholderia to demonstrate the physical kill-on-contact mechanism of action.
  • Time-kill resulted in an overall 4.44 log reduction in cfu/ml from the untreated beginning to the 30 minute time point.
  • Each 10 minute time point had between 1-2 log reduction as follows: from the untreated to the 10 minute point there was a 1.40 log reduction, from the 10 to 20 minute time points there was a 1.91 log reduction and from the 20 to 30 minute time points there was a 1.13 log reduction. See FIGS. 2A-2D .
  • nanoemulsions such as the tested P 407 5EC
  • the SEM images demonstrate the kill on contact mechanism, here in this case a Gram negative bacterium with a reputation of having a tough outer membrane. Further studies are ongoing to investigate the nebulization of P 407 5EC for the treatment of pulmonary infection in cystic fibrosis patients.
  • the present invention provides immunogenic compositions comprising a nanoemulsion and Burkholderia antigen (e.g., Burkholderia outer membrane protein (OMP)) that, when administered to a subject, induces immunity (e.g., protective immunity) in the subject against bacteria from the genus Burkholderia (e.g., B. cenocepacia, B. multivorans or others species associated with respiratory infection).
  • Burkholderia antigen e.g., Burkholderia outer membrane protein (OMP)
  • OMP outer membrane protein
  • the present invention provides all or a portion of an OMP (e.g., isolated, purified, and/or recombinant OMPs)) from a bacteria of the genus Burkholderia (See, e.g., Example 10, Table 25) that is utilized in an immunogenic composition comprising a nanoemulsion (e.g., for administration to a subject (e.g., to induce immunity to a bacteria of the genus Burkholderia in the subject)).
  • an immunogenic composition comprising a nanoemulsion and Burkholderia OMP induces both mucosal and systemic anti-OMP antibodies.
  • Example 10 the present invention provides that when administered to a subject, an immunogenic composition comprising a nanoemulsion and an OMP from a specific strain of Burkholderia (e.g., B. cenocepacia ) induces both mucosal and systemic anti-OMP antibodies that neutralize bacteria of the specific species from which the OMP is derived (e.g., B. cenocepacia ) and cross-neutralize bacteria from one or more other species of bacteria (e.g., of the genus Burkholderia ).
  • a specific strain of Burkholderia e.g., B. cenocepacia
  • mucosal and systemic anti-OMP antibodies that neutralize bacteria of the specific species from which the OMP is derived (e.g., B. cenocepacia ) and cross-neutralize bacteria from one or more other species of bacteria (e.g., of the genus Burkholderia ).
  • Example 10 shows that a single nasal immunization with OMP-NE produces a robust immune response characterized by rapid induction of antigen-specific T cell responses and secretory antibody responses, as documented by induction of IFN- ⁇ , IL-2 and mucosal sIgA and IgG, and that the immune response can be subsequently boosted.
  • Mice immunized with OMP-NE displayed a dramatic decrease in K56-1 colony forming units (cfu) in the lungs when compared with non-immunized controls. Pulmonary bacterial clearance was significantly enhanced in OMP-NE vaccinated mice which showed fewer B. cenocepacia organisms in the spleens as compared to non-vaccinated groups.
  • the present invention provides that mucosal OMP-NE administration produced protective immunity and reduced the likelihood of sepsis in subjects.
  • Example 10 also provides data identifying a 17 KDa OmpA-like protein and immune responses associated with exposure to the protein.
  • the 17 KDa lipoprotein fractions were better recognized by antibodies contained in serum from OMP-NE vaccinated mice (See, e.g., Example 10, FIG. 29C ).
  • the present invention provides that immunogenic compositions comprising epitopes contained within the 17 KDa OmpA-like proteins generate protective antibodies (e.g., against one or more species of Burkholderia ) in a subject administered the immunogenic composition.
  • OMP-NE and OMP in PBS responses were significantly differences between OMP-NE and OMP in PBS responses.
  • immunization with OMP in the absence of nanoemulsion resulted in relatively high titers of IgG, but were consistently lower (13 to 30 fold) than those induced by OMP-NE immunogenic compositions.
  • Immunization with OMP in the absence of nanoemulsion failed to respond to a booster immunization.
  • the response induced with OMP plus nanoemulsion was primarily directed toward specific proteins contained within the preparation, whereas OMP in the absence of nanoemulsion appears to have elicited immune responses to the intrinsic endotoxin content.
  • the present invention provides methods and compositions for the stimulation of immune responses.
  • the present invention provides immunogenic nanoemulsion compositions and methods of using the same for the induction of immune responses (e.g., innate and/or adaptive immune responses (e.g., for generation of host immunity against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans , etc.))).
  • immune responses e.g., innate and/or adaptive immune responses (e.g., for generation of host immunity against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans , etc.)
  • Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine (e.g., vaccination)) and research applications.
  • the present invention provides nanoemulsion adjuvants and compositions comprising the same (e.g., vaccines) for the stimulation of immune responses (e.g., immunity) against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans , etc.).
  • the present invention provides nanoemulsion adjuvant compositions that stimulate and/or elicit immune responses (e.g., innate immune responses and/or adaptive/acquired immune responses) when administered to a subject (e.g., a human or other mammalian subject)).
  • the present invention provides nanoemulsion adjuvant compositions comprising one or a plurality of Burkholderia (e.g., B. cenocepacia, B. multivorans , etc.) antigens (e.g., Burkholderia components isolated and/or purified, and/or recombinant Burkholderia proteins (e.g., OMPs)).
  • Burkholderia e.g., B. cenocepacia, B. multivorans , etc.
  • antigens e.g., Burkholderia components isolated and/or purified, and/or recombinant Burkholderia proteins (e.g., OMPs)
  • the present invention is not limited to any particular nanoemulsion or Burkholderia antigen.
  • Exemplary immunogenic compositions e.g., vaccine compositions
  • methods of administering the compositions are described in more detail below.
  • the present invention provides an immunogenic composition comprising a nanoemulsion and one or more Burkholderia antigens (e.g., B. cepacia antigens). In some embodiments, the present invention provides a method of inducing an immune response to Burkholderia (e.g., B.
  • cepacia in a subject comprising: providing a subject and an immunogenic composition comprising a nanoemulsion and an immunogen, wherein the immunogen comprises a Burkholderia (e.g., Burkholderia cepacia ) antigen and administering the composition to the subject under conditions such that the subject generates a Burkholderia (e.g., Burkholderia cepacia ) specific immune response.
  • the present invention is not limited by the route chosen for administration of a composition of the present invention.
  • administering the immunogenic composition comprises contacting a mucosal surface of the subject with the composition.
  • the mucosal surface comprises nasal mucosa.
  • inducing an immune response induces immunity to Burkholderia (e.g., Burkholderia cepacia ) in the subject.
  • a composition comprising a nanoemulsion (NE) and Burkholderia antigen could be utilized to generate an immune response in a subject.
  • Nasal immunization with a whole cell Streptococcus pneumoniae antigen (WCPAg) mixed with nanoemulsion was performed and shown to induce an IgG response in a host subject and the ability to eradicate upper respiratory colonization of S. pneumoniae.
  • the present invention provides immunogenic compositions comprising a nanoemulsion and Burkholderia antigen (e.g., Burkholderia outer membrane protein (OMP)) that, when administered to a subject, induces immunity (e.g., protective immunity) in the subject against bacteria from the genus Burkholderia (e.g., B. cenocepacia, B. multivorans or others species associated with respiratory infection).
  • a nanoemulsion and Burkholderia antigen e.g., Burkholderia outer membrane protein (OMP)
  • OMP outer membrane protein
  • the present invention provides that administration (e.g., nasal administration) of a composition comprising nanoemulsion and Burkholderia antigen (e.g., OMP antigen (e.g., a 17 KDa protein comprising an amino acid sequence selected from SEQ ID NOs. 1-16)) to a subject produces immunity toward Burkholderia in the subject thereby protecting the subject against Burkholderia infection (e.g., associated with a respiratory infection).
  • a composition comprising nanoemulsion and Burkholderia antigen
  • Burkholderia antigen e.g., OMP antigen (e.g., a 17 KDa protein comprising an amino acid sequence selected from SEQ ID NOs. 1-16)
  • OMP antigen e.g., a 17 KDa protein comprising an amino acid sequence selected from SEQ ID NOs. 1-16
  • the present invention is not limited by the type of bacteria of the genus Burkholderia utilized in the immunogenic compositions and methods of using the same of the invention.
  • the bacteria is a pathogen.
  • the pathogen is a Burkholderia species responsible for respiratory or respiratory associated infection.
  • a variety of Burkholderia species find use in the compositions and methods of the invention including, but not limited to, B. cenocepacia, B. dolosa, B. multivorans, B. ambifaria, B. vietnamiensis, B. ubonensis, B. thailandensis, B. graminis, B. oklahomensis, B. pseudomallei, B. xenovorans, B. phytofirmans, B. phymatum, R. metallidurans, R. eutropha, R. solanacearum.
  • the bacteria of the genus Burkholderia is B. cepecia .
  • an immunogenic composition comprising a nanoemulsion and an Omp-A like protein comprises an Omp-A like protein from a Burkholderia species including, but not limited to, B. cenocepacia, B. dolosa, B. multivorans, B. ambifaria, B. vietnamiensis, B. ubonensis, B. thailandensis, B. graminis, B. oklahomensis, B. pseudomallei, B. xenovorans, B. phytofirmans, B. phymatum, R. metallidurans, R. eutropha, R.
  • an immunogenic composition comprising a nanoemulsion and an Omp-A like protein comprises an Omp-A like protein (e.g., isolated, purified, and/or recombinant Omp-A like protein) comprising an amino acid sequence identified in SEQ ID NOs.: 1-16.
  • an immunogenic composition comprising a nanoemulsion and an Omp-A like protein comprises an Omp-A like protein (e.g., isolated, purified, and/or recombinant Omp-A like protein) comprising an amino acid sequence of SEQ ID NO. 1.
  • the number of Burkholderia antigens utilized can range from 2 different serotypes to about 20 different serotypes.
  • an immunogenic composition comprising a nanoemulsion and Burkholderia antigen may comprise Burkholderia antigen (e.g., whole cell, polysaccharide, Omp-A protein, other protein, etc.) from every known and/or isolated Burkholderia serotype.
  • Burkholderia antigen e.g., whole cell, polysaccharide, Omp-A protein, other protein, etc.
  • an immunogenic composition comprising a nanoemulsion and Burkholderia (e.g., B. cepacia ) antigen comprises one, two or more different types of carrier protein (e.g., that act as carriers for proteins, saccharides, etc.).
  • carrier protein e.g., that act as carriers for proteins, saccharides, etc.
  • two or more different saccharides or proteins may be conjugated to the same carrier protein, either to the same molecule of carrier protein or to different molecules of the same carrier protein.
  • Carrier proteins may be TT, DT, CRM197, fragment C of TT, PhtD, PhtBE or PhtDE fusions (particularly those described in WO 01/98334 and WO 03/54007), detoxified pneumolysin and protein D.
  • a carrier protein present in a composition comprising a nanoemulsion and Burkholderia (e.g., B. cenocepacia ) antigen is a member of the polyhistidine triad family (Pht) proteins, fragments or fusion proteins thereof.
  • the PhtA, PhtB, PhtD or PhtE proteins may have an amino acid sequence sharing 80%, 85%, 90%, 95%, 98%, 99% or 100% identity with a sequence disclosed in WO 00/37105 or WO 00/39299 (e.g. with amino acid sequence 1-838 or 21-838 of SEQ ID NO: 4 of WO 00/37105 for PhtD).
  • fusion proteins are composed of full length or fragments of 2, 3 or 4 of PhtA, PhtB, PhtD, PhtE.
  • fusion proteins are PhtA/B, PhtA/D, PhtA/E, PhtB/A, PhtB/D, PhtB/E. PhtD/A. PhtD/B, PhtD/E, PhtE/A, PhtE/B and PhtE/D, wherein the proteins are linked with the first mentioned at the N-terminus (see for example WO01/98334).
  • Carriers may comprise histidine triad motif(s) and/or coiled coil regions.
  • a histidine triad motif is the portion of polypeptide that has the sequence HxxHxH where H is histidine and x is an amino acid other than histidine.
  • a coiled coil region is a region predicted by “Coils” algorithm Lupus, A et al (1991) Science 252; 1162-1164.
  • carrier proteins which may be used in the present invention are DT (Diphtheria toxoid), TT (tetanus toxoid) or fragment C of TT, DT CRM197 (a DT mutant) other DT point mutants, such as CRM176, CRM228, CRM 45 (Uchida et al J. Biol. Chem.
  • meningitidis serogroup B EP0372501
  • PorB from N. meningitidis
  • PD Haemophilus influenzae protein D—see, e.g., EP 0 594 610 B), or immunologically functional equivalents thereof, synthetic peptides (EP0378881, EP0427347), heat shock proteins (WO 93/17712, WO 94/03208), pertussis proteins (WO 98/58668, EP0471177), cytokines, lymphokines, growth factors or hormones (WO 91/01146), artificial proteins comprising multiple human CD4+ T cell epitopes from various pathogen derived antigens (Falugi et al (2001) Eur J Immunol 31; 3816-3824) such as N19 protein (Baraldoi et al (2004) Infect Immun 72; 4884-7) pneumococcal surface protein PspA (WO 02/091998), iron uptake proteins (WO 01/72337), toxin A or B of C. difficile
  • An immunogenic composition comprising a nanoemulsion and Burkholderia (e.g., B. cenocepacia ) antigen can be used to immunize a mammal, such as a mouse, rat, rabbit, guinea pig, monkey, or human, to produce polyclonal antibodies.
  • a Burkholderia e.g., B. cenocepacia
  • a carrier protein such as bovine serum albumin, thyroglobulin, keyhole limpet hemocyanin or other carrier described herein.
  • various adjuvants can be used to increase the immunological response.
  • Such adjuvants include, but are not limited to, Freund's adjuvant, mineral gels (e.g., aluminum hydroxide), and surface active substances (e.g. lysolecithin, pluronic polyols, polyanions, peptides, nanoemulsions described herein, keyhole limpet hemocyanin, and dinitrophenol).
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are especially useful.
  • Monoclonal antibodies that specifically bind to a Burkholderia can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These techniques include, but are not limited to, the hybridoma technique, the human B cell hybridoma technique, and the EBV hybridoma technique (See, e.g., Kohler et al., Nature 256, 495 497, 1985; Kozbor et al., J. Immunol. Methods 81, 3142, 1985; Cote et al., Proc. Natl. Acad. Sci. 80, 2026 2030, 1983; Cole et al., Mol. Cell. Biol. 62, 109 120, 1984).
  • Burkholderia e.g., B. cepacia
  • chimeric antibodies the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used (See, e.g., Morrison et al., Proc. Natl. Acad. Sci. 81, 68516855, 1984; Neuberger et al., Nature 312, 604 608, 1984; Takeda et al., Nature 314, 452 454, 1985).
  • Monoclonal and other antibodies also can be “humanized” to prevent a patient from mounting an immune response against the antibody when it is used therapeutically.
  • Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between rodent antibodies and human sequences can be minimized by replacing residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grating of entire complementarity determining regions.
  • humanized antibodies can be produced using recombinant methods, as described below.
  • Antibodies which specifically bind to a particular antigen can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. Pat. No. 5,565,332.
  • single chain antibodies can be adapted using methods known in the art to produce single chain antibodies which specifically bind to a particular antigen.
  • Antibodies with related specificity, but of distinct idiotypic composition can be generated by chain shuffling from random combinatorial immunoglobin libraries (See, e.g., Burton, Proc. Natl. Acad. Sci. 88, 11120 23, 1991).
  • Single-chain antibodies also can be constructed using a DNA amplification method, such as PCR, using hybridoma cDNA as a template (See, e.g., Thirion et al., 1996, Eur. J. Cancer Prey. 5, 507-11).
  • Single-chain antibodies can be mono- or bispecific, and can be bivalent or tetravalent. Construction of tetravalent, bispecific single-chain antibodies is taught, for example, in Coloma & Morrison, 1997, Nat. Biotechnol. 15, 159-63. Construction of bivalent, bispecific single-chain antibodies is taught, for example, in Mallender & Voss, 1994, J. Biol. Chem. 269, 199-206.
  • a nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below.
  • single-chain antibodies can be produced directly using, for example, filamentous phage technology (See, e.g., Verhaar et al., 1995, Int. J. Cancer 61, 497-501; Nicholls et al., 1993, J. Immunol. Meth. 165, 81-91).
  • Antibodies which specifically bind to a particular antigen also can be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature (See, e.g., Orlandi et al., Proc. Natl. Acad. Sci. 86, 3833 3837, 1989; Winter et al., Nature 349, 293 299, 1991).
  • Chimeric antibodies can be constructed as disclosed in WO 93/03151. Binding proteins which are derived from immunoglobulins and which are multivalent and multispecific, such as the “diabodies” described in WO 94/13804, also can be prepared. Antibodies can be purified by methods well known in the art. For example, antibodies can be affinity purified by passage over a column to which the relevant antigen is bound. The bound antibodies can then be eluted from the column using a buffer with a high salt concentration.
  • Contemporary burn wound management involves early debridement and reconstruction of non-viable skin coupled with provision of supportive care and topical antimicrobial dressing changes to partial thickness burn wounds.
  • the goal of modern burn wound care is to provide an optimal environment for epidermal renewal. Restoration of skin integrity takes place via regrowth of keratinocytes from preserved hair follicles or transfer of split thickness skin grafts harvested from non-burn regions. During the period of epidermal renewal it is important to avoid further injury to the skin, abrogate burn wound progression, and minimize secondary complications such as wound infection.
  • topical antimicrobial agents include silver sulfadiazine (SILVADENE), mafenide acetate (SULFAMYLON), and colloidal silver impregnated dressings (ACTICOAT, SILVERLON).
  • Each of these agents has potential limitations such as variable ability to penetrate eschar, uneven efficacy against both Gram-negative and Gram-positive bacteria, and potential toxicity to host immune cells (See, e.g., Steinstraesser et al., Antimicrob Agents Chemother 46(6):1837-1844, 2002).
  • the present invention provides nanoemulsion compositions and methods of using the same for the treatment of burn wounds.
  • the present invention provides nanoemulsion compositions and methods of using the same to reduce, attenuate and/or prevent bacterial growth in a burn wound.
  • the present invention also provides nanoemulsion compositions that reduce wound inflammation following burn injury.
  • a nanoemulsion composition that is applied to a wound following burn injury is able to penetrate more deeply and uniformly into a burn wound (e.g., thereby decreasing and/or inhibiting bacterial growth and/or inflammation at the site of the wound).
  • the present invention provides a method of reducing, inhibiting and/or eliminating bacterial growth in a burn wound comprising providing a burn wound and a nanoemulsion and administering the nanoemulsion to the burn wound under conditions that bacterial growth is reduced, inhibited and/or eliminated.
  • a nanoemulsion composition described herein is combined with one or more antimicrobial drugs for administration to a burn wound to minimize bacterial growth at the burn wound site.
  • the present invention is not limited to any particular antimicrobial drug. Indeed, any antimicrobial drug that inhibits bacterial growth known to those in the art can be utilized in combination with a nanoemulsion composition described herein.
  • SIRS systemic inflammatory response syndrome
  • nanoemulsion compositions described herein can be administered to a burn wound to treat (e.g., reduce, attenuate and/or prevent) inflammation, tissue edema and/or intravascular hypovolemia at the site of a burn wound.
  • reducing inflammation, tissue edema and/or intravascular hypovolemia at the site of a burn wound reduces the occurrence of shock, pulmonary dysfunction, abdominal or extremity compartment syndrome, and/or cardiac failure.
  • a nanoemulsion composition described herein is used in combination with (e.g., is co-administered with) one or more anti-inflammatory drugs to minimize early burn wound inflammation and tissue edema.
  • the present invention is not limited to any particular anti-inflammatory drug. Indeed, any anti-inflammatory drug that minimizes early burn wound inflammation and tissue edema can be utilized in combination with a nanoemulsion composition described herein.
  • a nanoemulsion of the invention (e.g., W 20 5GBA 2 ED) is administered to a burn wound to prevent, attenuate and/or eradicate bacterial growth (e.g., Staphylococcus aureus, P. aeruginosa , or other bacteria) within a partial thickness burn wound.
  • bacterial growth e.g., Staphylococcus aureus, P. aeruginosa , or other bacteria
  • Example 9 shows that reduction in microbial infection was coupled with generation of lower levels of local dermal pro-inflammatory cytokines and evidence of reduced neutrophil sequestration into the burn wound. This decrease in burn wound bacterial growth and inflammation also produced less capillary leak in the early post-thermal injury time-period.
  • Topical antimicrobial therapy is used to reduce the microbial load in the burn wound and reduce this risk of infection.
  • Current topical agents include silver nitrate (AgNO 3 ), silver sulfadiazine, mafenide acetate, and nanocrystalline impregnated silver dressings.
  • Silver nitrate is limited in its use because of the problem it creates from contact staining and its limited antifungal activity.
  • Silver sulfadiazine is the mainstay of topical burn antimicrobial treatment. It is bactericidal against P.
  • aeruginosa and other Gram-negative enteric bacteria. Resistance to Silvadene by some of these organisms has emerged (See, e.g., Silver et al., J Ind Microbiol Biotechnol 33(7):627-634, 2006). The agent has limited antifungal activity, but can be used in conjunction with nystatin. Silvadene has no real ability to penetrate burn eschar and sometimes leads to leukopenia which requires conversion to another topical agent.
  • mafenide acetate is narrowed by the fact that it is bacteriostatic against select organisms, it has limited activity against Gram-positive bacteria such as Staphylococcus aureus , and that its use over a large surface area can lead to a metabolic acidosis because of its metabolism into a carbonic anhydrase inhibitor.
  • the nanocrystalline silver dressings have the broadest activity against burn wound pathogens of the current agents available. They have a modest ability to penetrate eschar and can be left in place for many days (See, e.g., Church et al., Clin Microbiol Rev 19(2):403-434, 2006).
  • SB 202190 an inhibitor of activated p38 MAPK, can substantially reduce the dermal inflammation generated in burn wounds (See, e.g., Arbabi et al., Shock. 26(2):201-209, 2006). Thermal injury initiates dermal inflammatory and pro-apoptotic cell signaling.
  • the present invention provides nanoemulsion compositions that can be utilized to reduce, when administered to a burn wound, conversion of the partial thickness burn wound within the “zone of stasis” to regions of full thickness burn.
  • the burn wound itself is the primary source triggering the systemic inflammatory response via generation of pro-inflammatory cytokines and sequestration of neutrophils into the burn wound (See, e.g., Hansbrough et al., J Surg Res 61(1):17-22, 1996; Piccolo et al., Inflammation 23(4):371-385, 1999; Till et al., J Clin Invest 69(5):1126-1135, 1982).
  • Topical application of a p38 MAPK inhibitor can control the source of inflammation at the level of the dermis, resulting in lower levels of pro-inflammatory mediators, reduced neutrophil sequestration and microvascular damage, and less epithelial apoptosis in burn wound hair follicle cells (See, e.g., Ipaktchi et al., Shock. 26(2):201-209, 2006).
  • Dermal source control of inflammation also reduces bacterial growth and attenuates the systemic inflammatory response resulting in less acute lung injury and cardiac dysfunction following partial thickness burn injury in a rodent model.
  • a nanoemulsion of the invention is utilized (e.g., administered) alone or in combination with an anti-inflammatory and/or antimicrobial agent to reduce local dermal inflammation and risk of infection within burn wounds (e.g., early burn wounds, partial thickness wounds, full thickness wounds or other burn wounds).
  • an anti-inflammatory and/or antimicrobial agent to reduce local dermal inflammation and risk of infection within burn wounds (e.g., early burn wounds, partial thickness wounds, full thickness wounds or other burn wounds).
  • the present invention is not limited by the type of anti-inflammatory agent and/or antimicrobial utilized for co-administration with a nanoemulsion described herein.
  • anti-inflammatory agents and/or antimicrobial agents can be used including, but not limited to, silver nitrate (AgNO 3 ), silver sulfadiazine, mafenide acetate, nanocrystalline impregnated silver dressings, p38 MAPK inhibitor (e.g., SB 202190), or another anti-inflammatory or antimicrobial agent described herein.
  • the nanoemulsion when a nanoemulsion of the invention is administered to a burn wound, can be administered (e.g., to a subject (e.g., to a burn or wound surface) by multiple methods, including, but not limited to, direct use or being suspended in a solution (e.g., colloidal solution) and applied to a surface (e.g., a surface comprising bacteria (e.g., pathogenic bacteria) or susceptible to bacterial invasion); being sprayed onto a surface using a spray applicator; being mixed with fibrin glue and applied (e.g., sprayed) onto a surface (e.g., skin burn or wound); being impregnated onto a wound dressing or bandage and applying the bandage to a surface (e.g., an infection or burn wound); being applied by a controlled-release mechanism; or being impregnated on one or both sides of an acellular biological matrix that is then placed on a surface (e.g., skin burn or wound) thereby protecting at
  • the invention provides a pharmaceutical composition containing (a) a composition comprising a nanoemulsion (e.g., W 20 5GBA 2 ED); and (b) one or more other agents (e.g., an antibiotic).
  • antibiotics include, but are not limited to, almecillin, amdinocillin, amikacin, amoxicillin, amphomycin, amphotericin B, ampicillin, azacitidine, azaserine, azithromycin, azlocillin, aztreonam, bacampicillin, bacitracin, benzyl penicilloyl-polylysine, bleomycin, candicidin, capreomycin, carbenicillin, cefaclor, cefadroxil, cefamandole, cefazoline, cefdinir, cefepime, cefixime, cefinenoxime, cefinetazole, cefodizime, cefonicid, cef
  • two or more combined agents may be used together or sequentially.
  • an antibiotic may comprise bacteriocins, type A lantibiotics, type B lantibiotics, liposidomycins, mureidomycins, alanoylcholines, quinolines, eveminomycins, glycylcyclines, carbapenems, cephalosporins, streptogramins, oxazolidonones, tetracyclines, cyclothialidines, bioxalomycins, cationic peptides, and/or protegrins.
  • the composition comprises lysostaphin.
  • the present invention is not limited by the type of nanoemulsion utilized (e.g., for respiratory administration, administration to a burn wound and/or for use in an immunogenic composition for induction of protective immune responses). Indeed, a variety of nanoemulsion compositions are contemplated to be useful in the present invention.
  • a nanoemulsion comprises (i) an aqueous phase; (ii) an oil phase; and at least one additional compound.
  • these additional compounds are admixed into either the aqueous or oil phases of the composition.
  • these additional compounds are admixed into a composition of previously emulsified oil and aqueous phases.
  • one or more additional compounds are admixed into an existing emulsion composition immediately prior to its use.
  • one or more additional compounds are admixed into an existing emulsion composition prior to the compositions immediate use.
  • Additional compounds suitable for use in a nanoemulsion of the present invention include, but are not limited to, one or more organic, and more particularly, organic phosphate based solvents, surfactants and detergents, cationic halogen containing compounds, germination enhancers, interaction enhancers, food additives (e.g., flavorings, sweeteners, bulking agents, and the like) and pharmaceutically acceptable compounds.
  • organic phosphate based solvents e.g., cationic halogen containing compounds
  • germination enhancers e.g., germination enhancers, interaction enhancers
  • food additives e.g., flavorings, sweeteners, bulking agents, and the like
  • pharmaceutically acceptable compounds e.g., exemplary embodiments of the various compounds contemplated for use in the compositions of the present invention are presented below. Unless described otherwise, nanoemulsions are described in undiluted form.
  • the nanoemulsions of the invention can be stable at about 40° C. and about 75% relative humidity for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, or at least up to about 3 years.
  • the nanoemulsions of the invention can be stable at about 25° C. and about 60% relative humidity for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, or at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, or at least up to about 5 years.
  • the nanoemulsions of the invention can be stable at about 4° C. for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, at least up to about 5 years, at least up to about 5.5 years, at least up to about 6 years, at least up to about 6.5 years, or at least up to about 7 years.
  • the nanoemulsions of the invention are stable upon application, as surprisingly the nanoemulsions do not lose their physical structure upon application.
  • Microscopic examination of skin surface following application of a nanoemulsion according to the invention demonstrates the physical integrity of the nanoemulsions of the invention. This physical integrity may result in the desired absorption observed with the nanoemulsions of the invention.
  • nanoemulsion refers to a dispersion or droplet or any other lipid structure.
  • Typical lipid structures contemplated in the invention include, but are not limited to, unilamellar, paucilamellar and multilamellar lipid vesicles, micelles and lamellar phases.
  • the nanoemulsion of the present invention comprises droplets having an average diameter size of less than about 1,000 nm, less than about 950 nm, less than about 900 nm, less than about 850 nm, less than about 800 nm, less than about 750 nm, less than about 700 nm, less than about 650 nm, less than about 600 nm, less than about 550 nm, less than about 500 nm, less than about 450 nm, less than about 400 nm, less than about 350 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, or any combination thereof.
  • the droplets have an average diameter size greater than about 125 nm and less than or equal to about 300 nm. In a different embodiment, the droplets have an average diameter size greater than about 50 nm or greater than about 70 nm, and less than or equal to about 125 nm. In other embodiments of the invention, the nanoemulsion droplets have an average diameter of from about 300 nm to about 600 nm; or the nanoemulsion droplets have an average diameter of from about 150 nm to about 400 nm.
  • the nanoemulsion has a narrow range of MIC (minimum inhibitory concentration) and MBC (minimum bactericidal concentrations) values.
  • the MIC and MBC for the nanoemulsion differ by less than or equal to four-fold, meaning that the nanoemulsion is bactericidal.
  • the MIC and MBC for the nanoemulsion may differ by greater than four-fold, meaning that the nanoemulsion is bacteriostatic.
  • the nanoemulsion comprises: (a) an aqueous phase; (b) about 1% oil to about 80% oil; (c) about 0.1% organic solvent to about 50% organic solvent; (d) about 0.001% surfactant or detergent to about 10% surfactant or detergent; (e) about 0.0005% to about 1.0% of a chelating agent; or (e) any combination thereof.
  • the nanoemulsion comprises: (a) about 10% oil to about 80% oil; (b) about 1% organic solvent to about 50% organic solvent; (c) at least one non-ionic surfactant present in an amount of about 0.1% to about 10%; (d) at least one cationic agent present in an amount of about 0.01% to about 3%; or any combination thereof.
  • the nanoemulsion comprises a cationic surfactant which is either cetylpyridinium chloride (CPC) or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or combination thereof.
  • a cationic surfactant which is either cetylpyridinium chloride (CPC) or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or combination thereof.
  • the cationic surfactant may have a concentration in the nanoemulsion of less than about 5.0% and greater than about 0.001%, or further, may have a concentration of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, less than about 0.10%, greater than about 0.001%, greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, and greater than about 0.010%.
  • the nanoemulsion comprises a non-ionic surfactant, and may have a concentration of about 0.01% to about 10.0%, or about 0.1% to about 3% of a non-ionic surfactant, such as a polysorbate.
  • the nanoemulsion (a) comprises at least one cationic surfactant; (b) comprises a cationic surfactant which is either cetylpyridinium chloride or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or combination thereof; (c) comprises a cationic surfactant, and wherein the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001%; (d) comprises a cationic surfactant, and wherein the concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%
  • the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant;
  • the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant;
  • the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a polysorbate nonionic surfactant;
  • the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant, and the non-ionic surfactant is present in a concentration of about 0.05% to about 10%, about 0.05% to about 7.0%, about 0.1% to about 7%, or about 0.5% to about 5%;
  • the nanoemulsion comprises at least one cationic surfactant
  • the nanoemulsion comprises: (a) water; (b) ethanol or glycerol (glycerine), or a combination thereof; (c) either cetylpyridinium chloride (CPC), or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or a combination thereof; (c) soybean oil; and (e) Poloxamer 407, Tween 80, or Tween 20.
  • the nanoemulsion can further comprise EDTA.
  • nanoemulsions tested in vitro generally have lower concentrations of oil, organic solvent, surfactant or detergent, and (if present) chelating agent than that present in a nanoemulsion intended for therapeutic use, e.g., topical use. This is because in vitro studies do not require the nanoemulsion droplets to traverse the skin. For topical, aerosol, intradermal etc. use, the concentrations of the components must be higher to result in a therapeutic nanoemulsion.
  • the relative quantities of each component used in a nanoemulsion tested in vitro are applicable to a nanoemulsion to be used therapeutically and, therefore, in vitro quantities can be scaled up to prepare a therapeutic composition, and in vitro data is predictive of topical application success.
  • the aqueous phase can comprise any type of aqueous phase including, but not limited to, water (e.g., H 2 O, distilled water, tap water) and solutions (e.g., phosphate buffered saline (PBS) solution).
  • the aqueous phase comprises water at a pH of about 4 to 10, preferably about 6 to 8.
  • the water can be deionized (hereinafter “DiH 2 O”).
  • the aqueous phase comprises phosphate buffered saline (PBS).
  • the aqueous phase may further be sterile and pyrogen free.
  • Organic solvents in the nanoemulsions of the invention include, but are not limited to, C 1 -C 12 alcohol, diol, triol, dialkyl phosphate, tri-alkyl phosphate, such as tri-n-butyl phosphate, semi-synthetic derivatives thereof, and combinations thereof.
  • the organic solvent is an alcohol chosen from a nonpolar solvent, a polar solvent, a protic solvent, or an aprotic solvent.
  • Suitable organic solvents for the nanoemulsion include, but are not limited to, ethanol, methanol, isopropyl alcohol, glycerol, medium chain triglycerides, diethyl ether, ethyl acetate, acetone, dimethyl sulfoxide (DMSO), acetic acid, n-butanol, butylene glycol, perfumers alcohols, isopropanol, n-propanol, formic acid, propylene glycols, glycerol, sorbitol, industrial methylated spirit, triacetin, hexane, benzene, toluene, diethyl ether, chloroform, 1,4-dixoane, tetrahydrofuran, dichloromethane, acetone, acetonitrile, dimethylformamide, dimethyl sulfoxide, formic acid, semi-synthetic derivatives thereof, and any combination thereof.
  • DMSO dimethyl
  • the oil in the nanoemulsion of the invention can be any cosmetically or pharmaceutically acceptable oil.
  • the oil can be volatile or non-volatile, and may be chosen from animal oil, vegetable oil, natural oil, synthetic oil, hydrocarbon oils, silicone oils, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable oils include, but are not limited to, mineral oil, squalene oil, flavor oils, silicon oil, essential oils, water insoluble vitamins, Isopropyl stearate, Butyl stearate, Octyl palmitate, Cetyl palmitate, Tridecyl behenate, Diisopropyl adipate, Dioctyl sebacate, Menthyl anthranhilate, Cetyl octanoate, Octyl salicylate, Isopropyl myristate, neopentyl glycol dicarpate cetols, Ceraphyls®, Decyl oleate, diisopropyl adipate, C 12-15 alkyl lactates, Cetyl lactate, Lauryl lactate, Isostearyl neopentanoate, Myristyl lactate, Isocetyl stearoyl stearate, Octyldodecyl stearoyl
  • the oil may further comprise a silicone component, such as a volatile silicone component, which can be the sole oil in the silicone component or can be combined with other silicone and non-silicone, volatile and non-volatile oils.
  • Suitable silicone components include, but are not limited to, methylphenylpolysiloxane, simethicone, dimethicone, phenyltrimethicone (or an organomodified version thereof), alkylated derivatives of polymeric silicones, cetyl dimethicone, lauryl trimethicone, hydroxylated derivatives of polymeric silicones, such as dimethiconol, volatile silicone oils, cyclic and linear silicones, cyclomethicone, derivatives of cyclomethicone, hexamethylcyclotrisiloxane, octamethylcyclotetrasiloxane, decamethylcyclopentasiloxane, volatile linear dimethylpolysiloxanes, isohexadecane, is
  • the volatile oil can be the organic solvent, or the volatile oil can be present in addition to an organic solvent.
  • Suitable volatile oils include, but are not limited to, a terpene, monoterpene, sesquiterpene, carminative, azulene, menthol, camphor, thujone, thymol, nerol, linalool, limonene, geraniol, perillyl alcohol, nerolidol, farnesol, y GmbHe, bisabolol, farnesene, ascaridole, chenopodium oil, citronellal, citral, citronellol, chamazulene, yarrow, guaiazulene, chamomile, semi-synthetic derivatives, or combinations thereof.
  • the volatile oil in the silicone component is different than the oil in the oil phase.
  • the surfactant or detergent in the nanoemulsion of the invention can be a pharmaceutically acceptable ionic surfactant, a pharmaceutically acceptable nonionic surfactant, a pharmaceutically acceptable cationic surfactant, a pharmaceutically acceptable anionic surfactant, or a pharmaceutically acceptable zwitterionic surfactant.
  • Exemplary useful surfactants are described in Applied Surfactants: Principles and Applications. Tharwat F. Tadros, Copyright 8 2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-30629-3), which is specifically incorporated by reference.
  • the surfactant can be a pharmaceutically acceptable ionic polymeric surfactant, a pharmaceutically acceptable nonionic polymeric surfactant, a pharmaceutically acceptable cationic polymeric surfactant, a pharmaceutically acceptable anionic polymeric surfactant, or a pharmaceutically acceptable zwitterionic polymeric surfactant.
  • polymeric surfactants include, but are not limited to, a graft copolymer of a poly(methyl methacrylate) backbone with multiple (at least one) polyethylene oxide (PEO) side chain, polyhydroxystearic acid, an alkoxylated alkyl phenol formaldehyde condensate, a polyalkylene glycol modified polyester with fatty acid hydrophobes, a polyester, semi-synthetic derivatives thereof, or combinations thereof.
  • PEO polyethylene oxide
  • Surface active agents or surfactants are amphipathic molecules that consist of a non-polar hydrophobic portion, usually a straight or branched hydrocarbon or fluorocarbon chain containing 8-18 carbon atoms, attached to a polar or ionic hydrophilic portion.
  • the hydrophilic portion can be nonionic, ionic or zwitterionic.
  • the hydrocarbon chain interacts weakly with the water molecules in an aqueous environment, whereas the polar or ionic head group interacts strongly with water molecules via dipole or iondipole interactions.
  • surfactants are classified into anionic, cationic, zwitterionic, nonionic and polymeric surfactants.
  • Suitable surfactants include, but are not limited to, ethoxylated nonylphenol comprising 9 to 10 units of ethyleneglycol, ethoxylated undecanol comprising 8 units of ethyleneglycol, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate, polyoxyethylene (20) sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monostearate, sorbitan monooleate, ethoxylated hydrogenated ricin oils, sodium laurylsulfate, a diblock copolymer of ethyleneoxyde and propyleneoxyde, Ethylene Oxide-Propylene Oxide Block Copolymers, and tetra-functional block copolymers based on ethylene oxide and propylene oxide, Glyceryl monoesters, Glyceryl caprate, Glyceryl cap
  • Additional suitable surfactants include, but are not limited to, non-ionic lipids, such as glyceryl laurate, glyceryl myristate, glyceryl dilaurate, glyceryl dimyristate, semi-synthetic derivatives thereof, and mixtures thereof.
  • non-ionic lipids such as glyceryl laurate, glyceryl myristate, glyceryl dilaurate, glyceryl dimyristate, semi-synthetic derivatives thereof, and mixtures thereof.
  • the surfactant is a polyoxyethylene fatty ether having a polyoxyethylene head group ranging from about 2 to about 100 groups, or an alkoxylated alcohol having the structure R 5 —(OCH 2 CH 2 ) y —OH, wherein R 5 is a branched or unbranched alkyl group having from about 6 to about 22 carbon atoms and y is between about 4 and about 100, and preferably, between about 10 and about 100.
  • the alkoxylated alcohol is the species wherein R 5 is a lauryl group and y has an average value of 23.
  • the surfactant is an alkoxylated alcohol which is an ethoxylated derivative of lanolin alcohol.
  • the ethoxylated derivative of lanolin alcohol is laneth-10, which is the polyethylene glycol ether of lanolin alcohol with an average ethoxylation value of 10.
  • Nonionic surfactants include, but are not limited to, an ethoxylated surfactant, an alcohol ethoxylated, an alkyl phenol ethoxylated, a fatty acid ethoxylated, a monoalkaolamide ethoxylated, a sorbitan ester ethoxylated, a fatty amino ethoxylated, an ethylene oxide-propylene oxide copolymer, Bis(polyethylene glycol bis[imidazoyl carbonyl]), nonoxynol-9, Bis(polyethylene glycol bis[imidazoyl carbonyl]), Brij® 35, Brij® 56, Brij® 72, Brij® 76, Brij® 92V, Brij® 97, Brij® 58P, Cremophor® EL, Decaethylene glycol monododecyl ether, N-Decanoyl-N-methylglucamine, n-Decyl alpha-D-glucopyran
  • the nonionic surfactant can be a poloxamer.
  • Poloxamers are polymers made of a block of polyoxyethylene, followed by a block of polyoxypropylene, followed by a block of polyoxyethylene.
  • the average number of units of polyoxyethylene and polyoxypropylene varies based on the number associated with the polymer. For example, the smallest polymer, Poloxamer 101, consists of a block with an average of 2 units of polyoxyethylene, a block with an average of 16 units of polyoxypropylene, followed by a block with an average of 2 units of polyoxyethylene.
  • Poloxamers range from colorless liquids and pastes to white solids.
  • Poloxamers are used in the formulation of skin cleansers, bath products, shampoos, hair conditioners, mouthwashes, eye makeup remover and other skin and hair products.
  • Examples of Poloxamers include, but are not limited to, Poloxamer 101, Poloxamer 105, Poloxamer 108, Poloxamer 122, Poloxamer 123, Poloxamer 124, Poloxamer 181, Poloxamer 182, Poloxamer 183, Poloxamer 184, Poloxamer 185, Poloxamer 188, Poloxamer 212, Poloxamer 215, Poloxamer 217, Poloxamer 231, Poloxamer 234, Poloxamer 235, Poloxamer 237, Poloxamer 238, Poloxamer 282, Poloxamer 284, Poloxamer 288, Poloxamer 331, Poloxamer 333, Poloxamer 334, Poloxamer 335, Poloxamer 338, Poloxamer 401,
  • Suitable cationic surfactants include, but are not limited to, a quarternary ammonium compound, an alkyl trimethyl ammonium chloride compound, a dialkyl dimethyl ammonium chloride compound, a cationic halogen-containing compound, such as cetylpyridinium chloride, Benzalkonium chloride, Benzalkonium chloride, Benzyldimethylhexadecylammonium chloride, Benzyldimethyltetradecylammonium chloride, Benzyldodecyldimethylammonium bromide, Benzyltrimethylammonium tetrachloroiodate, Dimethyldioctadecylammonium bromide, Dodecylethyldimethylammonium bromide, Dodecyltrimethylammonium bromide, Dodecyltrimethylammonium bromide, Ethylhexadecyldimethylammonium
  • Exemplary cationic halogen-containing compounds include, but are not limited to, cetylpyridinium halides, cetyltrimethylammonium halides, cetyldimethylethylammonium halides, cetyldimethylbenzylammonium halides, cetyltributylphosphonium halides, dodecyltrimethylammonium halides, or tetradecyltrimethylammonium halides.
  • suitable cationic halogen containing compounds comprise, but are not limited to, cetylpyridinium chloride (CPC), cetyltrimethylammonium chloride, cetylbenzyldimethylammonium chloride, cetylpyridinium bromide (CPB), cetyltrimethylammonium bromide (CTAB), cetyidimethylethylammonium bromide, cetyltributylphosphonium bromide, dodecyltrimethylammonium bromide, and tetrad ecyltrimethylammonium bromide.
  • the cationic halogen containing compound is CPC, although the compositions of the present invention are not limited to formulation with a particular cationic containing compound.
  • Suitable anionic surfactants include, but are not limited to, a carboxylate, a sulphate, a sulphonate, a phosphate, chenodeoxycholic acid, chenodeoxycholic acid sodium salt, cholic acid, ox or sheep bile, Dehydrocholic acid, Deoxycholic acid, Deoxycholic acid, Deoxycholic acid methyl ester, Digitonin, Digitoxigenin, N,N-Dimethyldodecylamine N-oxide, Docusate sodium salt, Glycochenodeoxycholic acid sodium salt, Glycocholic acid hydrate, synthetic, Glycocholic acid sodium salt hydrate, synthetic, Glycodeoxycholic acid monohydrate, Glycodeoxycholic acid sodium salt, Glycolithocholic acid 3-sulfate disodium salt, Glycolithocholic acid ethyl ester, N-Lauroylsarcosine sodium salt, N-Lauroylsarcos
  • Suitable zwitterionic surfactants include, but are not limited to, an N-alkyl betaine, lauryl amindo propyl dimethyl betaine, an alkyl dimethyl glycinate, an N-alkyl amino propionate, CHAPS, minimum 98% (TLC), CHAPS, SigmaUltra, minimum 98% (TLC), CHAPS, for electrophoresis, minimum 98% (TLC), CHAPSO, minimum 98%, CHAPSO, SigmaUltra, CHAPSO, for electrophoresis, 3-(Decyldimethylammonio)propanesulfonate inner salt, 3-Dodecyldimethylammonio)propanesulfonate inner salt, SigmaUltra, 3-(Dodecyldimethylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylmyristylammonio)propanesulfonate, 3-(N,N-Dimethyloctadecylam
  • the nanoemulsion comprises a cationic surfactant, which can be cetylpyridinium chloride. In other embodiments of the invention, the nanoemulsion comprises a cationic surfactant, and the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001%.
  • the nanoemulsion comprises a cationic surfactant
  • concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, or less than about 0.10%.
  • the concentration of the cationic agent in the nanoemulsion is greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, greater than about 0.010%, or greater than about 0.001%. In one embodiment, the concentration of the cationic agent in the nanoemulsion is less than about 5.0% and greater than about 0.001%.
  • the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant.
  • the non-cationic surfactant is a nonionic surfactant, such as a polysorbate (Tween), such as polysorbate 80 or polysorbate 20.
  • the non-ionic surfactant is present in a concentration of about 0.05% to about 7.0%, or the non-ionic surfactant is present in a concentration of about 0.5% to about 4%.
  • the nanoemulsion comprises a cationic surfactant present in a concentration of about 0.01% to about 2%, in combination with a nonionic surfactant.
  • Additional compounds suitable for use in the nanoemulsions of the invention include but are not limited to one or more solvents, such as an organic phosphate-based solvent, bulking agents, coloring agents, pharmaceutically acceptable excipients, a preservative, pH adjuster, buffer, chelating agent, etc.
  • the additional compounds can be admixed into a previously emulsified nanoemulsion, or the additional compounds can be added to the original mixture to be emulsified.
  • one or more additional compounds are admixed into an existing nanoemulsion composition immediately prior to its use.
  • Suitable preservatives in the nanoemulsions of the invention include, but are not limited to, cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha-tocophernol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, sodium ascorbate, sodium metabisulphite, citric acid, edetic acid, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable preservatives include, but are not limited to, benzyl alcohol, chlorhexidine (bis(p-chlorophenyldiguanido) hexane), chlorphenesin (3-(-4-chloropheoxy)-propane-1,2-diol), Kathon CG (methyl and methylchloroisothiazolinone), parabens (methyl, ethyl, propyl, butyl hydrobenzoates), phenoxyethanol (2-phenoxyethanol), sorbic acid (potassium sorbate, sorbic acid), Phenonip (phenoxyethanol, methyl, ethyl, butyl, propyl parabens), Phenoroc (phenoxyethanol 0.73%, methyl paraben 0.2%, propyl paraben 0.07%), Liquipar Oil (isopropyl, isobutyl, butylparabens), Liquipar PE (70% phenoxyethanol, 30% liquipar oil), Nip
  • the nanoemulsion may further comprise at least one pH adjuster.
  • pH adjusters in the nanoemulsion of the invention include, but are not limited to, diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof.
  • the nanoemulsion can comprise a chelating agent.
  • the chelating agent is present in an amount of about 0.0005% to about 1.0%.
  • chelating agents include, but are not limited to, phytic acid, polyphosphoric acid, citric acid, gluconic acid, acetic acid, lactic acid, ethylenediamine, ethylenediaminetetraacetic acid (EDTA), and dimercaprol, and a preferred chelating agent is ethylenediaminetetraacetic acid.
  • the nanoemulsion can comprise a buffering agent, such as a pharmaceutically acceptable buffering agent.
  • buffering agents include, but are not limited to, 2-Amino-2-methyl-1,3-propanediol, ⁇ 9.5% (NT), 2-Amino-2-methyl-1-propanol, ⁇ 99.0% (GC), L-(+)-Tartaric acid, ⁇ 99.5% (T), ACES, ⁇ 99.5% (T), ADA, ⁇ 99.0% (T), Acetic acid, ⁇ 09.5% (GC/T), Acetic acid, for luminescence, ⁇ 99.5% (GC/T), Ammonium acetate solution, for molecular biology, ⁇ 5 M in H 2 O, Ammonium acetate, for luminescence, ⁇ 99.0% (calc.
  • KT Citrate Concentrated Solution, for molecular biology, 1 M in H 2 O, Citric acid, anhydrous, ⁇ 99.5% (T), Citric acid, for luminescence, anhydrous, ⁇ 99.5% (T), Diethanolamine, ⁇ 99.5% (GC), EPPS, ⁇ 99.0% (T), Ethylenediaminetetraacetic acid disodium salt dihydrate, for molecular biology, ⁇ 99.0% (T), Formic acid solution, 1.0 M in H 2 O, Gly-Gly-Gly, ⁇ 99.0% (NT), Gly-Gly, ⁇ 99.5% (NT), Glycine, ⁇ 99.0% (NT), Glycine, for luminescence, ⁇ 99.0% (NT), Glycine, for molecular biology, ⁇ 99.0% (NT), HEP
  • KT Magnesium formate solution, 0.5 M in H 2 O, Magnesium phosphate dibasic trihydrate, ⁇ 98.0%
  • KT Neutralization solution for the in-situ hybridization for in-situ hybridization, for molecular biology, Oxalic acid dihydrate, ⁇ 99.5% (RT), PIPES, ⁇ 99.5% (T), PIPES, for molecular biology, ⁇ 99.5% (T), Phosphate buffered saline, solution (autoclaved), Phosphate buffered saline, washing buffer for peroxidase conjugates in Western Blotting, 10 ⁇ concentrate, piperazine, anhydrous, ⁇ 99.0% (T), Potassium D-tartrate monobasic, ⁇ 99.0% (T), Potassium acetate solution, for molecular biology, Potassium acetate solution, for molecular biology, 5 M in H 2 O, Potassium acetate solution, for molecular biology
  • T Sodium citrate monobasic, anhydrous, ⁇ 99.5% (T), Sodium citrate tribasic dihydrate, ⁇ 99.0% (NT), Sodium citrate tribasic dihydrate, for luminescence, ⁇ 99.0% (NT), Sodium citrate tribasic dihydrate, for molecular biology, ⁇ 99.5% (NT), Sodium formate solution, 8 M in H 2 O, Sodium oxalate, ⁇ 99.5% (RT), Sodium phosphate dibasic dihydrate, ⁇ 99.0% (T), Sodium phosphate dibasic dihydrate, for luminescence, ⁇ 99.0% (T), Sodium phosphate dibasic dihydrate, for molecular biology, ⁇ 99.0% (T), Sodium phosphate dibasic dodecahydrate, ⁇ 99.0% (T), Sodium phosphate dibasic solution, 0.5 M in H 2 O, Sodium phosphate dibasic, anhydrous, ⁇ 99.5% (T), Sodium phosphate dibasic solution,
  • T TM buffer solution, for molecular biology, pH 7.4, TNT buffer solution, for molecular biology, pH 8.0, TRIS Glycine buffer solution, 10 ⁇ concentrate, TRIS acetate—EDTA buffer solution, for molecular biology, TRIS buffered saline, 10 ⁇ concentrate, TRIS glycine SDS buffer solution, for electrophoresis, 10 ⁇ concentrate, TRIS phosphate—EDTA buffer solution, for molecular biology, concentrate, 10 ⁇ concentrate, Tricine, ⁇ 99.5% (NT), Triethanolamine, ⁇ 99.5% (GC), Triethylamine, ⁇ 99.5% (GC), Triethylammonium acetate buffer, volatile buffer, ⁇ 1.0 M in H 2 O, Triethylammonium phosphate solution, volatile buffer, ⁇ 1.0 M in H 2 O, Trimethylammonium acetate solution, volatile buffer, ⁇ 1.0 M in H 2 O, Trimethylammonium phosphate solution, volatile buffer, ⁇ 1 M in H 2
  • the nanoemulsion can comprise one or more emulsifying agents to aid in the formation of emulsions.
  • Emulsifying agents include compounds that aggregate at the oil/water interface to form a kind of continuous membrane that prevents direct contact between two adjacent droplets.
  • Certain embodiments of the present invention feature nanoemulsions that may readily be diluted with water to a desired concentration without impairing their antiviral properties.
  • a nanoemulsion comprises an additional active agent, such as an antibiotic or a palliative agent (such as for burn wound treatment). Addition of another agent may enhance the therapeutic effectiveness of the nanoemulsion.
  • the nanoemulsion in and of itself has anti-bacterial activity and does not need to be combined with another active agent to obtain therapeutic effectiveness. Any antibacterial (or antibiotic) agent suitable for treating a bacterial infection can be incorporated into the topical nanoemulsions of the invention.
  • antibiotic agents include, but are not limited to, aminoglycosides, Ansamycins, Carbacephems, Carbapenems, Cephalosporins, Glycopeptides, Macrolides, Monobactams, Penicillins, Polypeptides, Polymyxin, Quinolones, Sulfonamides, Tetracyclines, and others (e.g., Arsphenamine, Chloramphenicol, Clindamycin, Lincomycin, Ethambutol, Fosfomycin, Fusidic acid, Furazolidone, Isoniazid, Linezolid, Metronidazole, Mupirocin, Nitrofurantoin, Platensimycin, Pyrazinamide, Quinupristin/Dalfopristin, Rifampicin (Rifampin in US), Thiamphenicol, Timidazole, Dapsone, and lofazimine).
  • Arsphenamine Chloramphenicol, Clindamycin, Lin
  • antibiotics examples include, but are not limited to, Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Streptomycin, Tobramycin, Paromomycin, Geldanamycin, Herbimycin, Loracarbef, Ertapenem, Doripenem, Imipenem/Cilastatin, Meropenem, Cefadroxil, Cefazolin, Cefalotin or Cefalothin, Cefalexin, Cefaclor, Cefamandole, Cefoxitin, Cefprozil, Cefuroxime, Cefixime, Cefdinir, Cefditoren, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefepime, Ceftobiprole, Teicoplanin, Vancomycin, Azithromycin, Clarithromycin, Dirithromycin, Ery
  • palliative agents which may be incorporated into the nanoemulsions of the invention include, but are not limited to, menthol, camphor, phenol, allantoin, benzocaine, corticosteroids, phenol, zinc oxide, camphor, pramoxine, dimethicone, meradimate, octinoxate, octisalate, oxybenzone, dyclonine, alcohols (e.g., benzyl alcohol), mineral oil, propylene glycol, titanium dioxide, silver nitrate (AgNO 3 ), silver sulfadiazine, mafenide acetate, nanocrystalline impregnated silver dressings, a p38 MAPK inhibitor, and magnesium stearate.
  • alcohols e.g., benzyl alcohol
  • mineral oil propylene glycol
  • titanium dioxide silver nitrate (AgNO 3 )
  • silver sulfadiazine mafenide acetate
  • nanoemulsions of the invention may be formulated into pharmaceutical compositions that comprise the nanoemulsion in a therapeutically effective amount and suitable, pharmaceutically-acceptable excipients for administration to a human subject in need thereof using any conventional pharmaceutical method of administration.
  • excipients are well known in the art.
  • terapéuticaally effective amount it is meant any amount of the nanoemulsion that is effective in treating microorganisms by killing or inhibiting the growth of the microorganisms, causing the microorganisms to lose pathogenicity, or any combination thereof.
  • Exemplary dosage forms may include, but are not limited to, patches, ointments, creams, emulsions, liquids, lotions, gels, bioadhesive gels, aerosols, pastes, foams, sunscreens, capsules, microcapsules, or in the form of an article or carrier, such as a bandage, insert, syringe-like applicator, pessary, powder, and talc or other solid.
  • the pharmaceutical compositions may be formulated for immediate release, sustained release, controlled release, delayed release, or any combinations thereof.
  • the formulations may comprise a penetration-enhancing agent for enhancing penetration of the nanoemulsion through the stratum corneum and into the epidermis or dermis (i.e., for methods of treating burn wounds).
  • Suitable penetration-enhancing agents include, but are not limited to, alcohols such as ethanol, triglycerides and aloe compositions.
  • the amount of the penetration-enhancing agent may comprise from about 0.5% to about 40% by weight of the formulation.
  • the nanoemulsions of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis.
  • electrophoretic delivery/electrophoresis Such transdermal methods, which comprise applying an electrical current, are well known in the art.
  • minimal systemic absorption of the nanoemulsion occurs upon topical administration.
  • Such minimal systemic exposure can be determined by the detection of less than 10 ng/mL, less than 8 ng/mL, less than 5 ng/mL, less than 4 ng/mL, less than 3 ng/mL, or less than 2 ng/mL of the one or more surfactants present in the nanoemulsion in the plasma of the subject.
  • Lack of systemic absorption may be monitored, for example, by measuring the amount of the surfactant, such as the cationic surfactant, in the plasma of the human subject undergoing treatment. Amounts of surfactant of equal to or less than about 10 ng/ml in the plasma confirms minimal systemic absorption.
  • the pharmaceutical compositions may be applied in a single administration or in multiple administrations.
  • the pharmaceutical compositions can be applied for at least one day, at least two days at least three days at least four days at least 5 days, once a week, at least twice a week, at least once a day, at least twice a day, multiple times daily, multiple times weekly, biweekly, at least once a month, or any combination thereof.
  • the nanoemulsion may be occluded or semi-occluded. Occlusion or semi-occlusion may be performed by overlaying a bandage, polyolefin film, article of clothing, impermeable barrier, or semi-impermeable barrier to the topical preparation.
  • CPC cetylpyridinium chloride, which is a cationic surfactant present in the nanoemulsions.
  • Compositions are w/w % unless otherwise noted.
  • the following nanoemulsions have an average particle (droplet) size of about 300 nm to about 600 nm: W 20 5EC ED, P 407 5EC, W 20 5GBA 2 , W 80 5EC, and W 20 5GBA 2 ED.
  • the W 20 5ECEDL2 which undergoes high pressure processing has an average particle (droplet) size of about 150 nm to about 400 nm.
  • the formulations listed in the table above are “neat” or “concentrated” formulations, meaning that the formulation intended for therapeutic use can be diluted as desired.
  • the nanoemulsions of the invention can be formed using classic emulsion forming techniques. See e.g., U.S. 2004/0043041. See also U.S. Pat. Nos. 6,015,832, 6,506,803, 6,559,189, 6,635,676, and US Patent Publication No. 20040043041, all of which are incorporated by reference. In addition, methods of making emulsions are described in U.S. Pat. Nos. 5,103,497 and 4,895,452 (herein incorporated by reference).
  • the oil is mixed with the aqueous phase under relatively high shear forces (e.g., using high hydraulic and mechanical forces) to obtain a nanoemulsion comprising oil droplets having an average diameter of less than about 1000 nm.
  • relatively high shear forces e.g., using high hydraulic and mechanical forces
  • Some embodiments of the invention employ a nanoemulsion having an oil phase comprising an alcohol such as ethanol.
  • the oil and aqueous phases can be blended using any apparatus capable of producing shear forces sufficient to form an emulsion, such as French Presses or high shear mixers (e.g., FDA approved high shear mixers are available, for example, from Admix, Inc., Manchester, N.H.). Methods of producing such emulsions are described in U.S. Pat. Nos. 5,103,497 and 4,895,452, herein incorporated by reference in their entireties.
  • the nanoemulsions used in the methods of the invention comprise droplets of an oily discontinuous phase dispersed in an aqueous continuous phase, such as water.
  • the nanoemulsions of the invention are stable, and do not decompose even after long storage periods. Certain nanoemulsions of the invention are non-toxic and safe when swallowed, inhaled, or contacted to the skin of a subject.
  • compositions of the invention can be produced in large quantities and are stable for many months at a broad range of temperatures.
  • the nanoemulsion can have textures ranging from that of a semi-solid cream to that of a thin lotion, and can be applied topically by hand, and can be sprayed onto a surface or nebulized.
  • the emulsion may be in the form of lipid structures including, but not limited to, unilamellar, multilamellar, and paucliamellar lipid vesicles, micelles, and lamellar phases.
  • the present invention contemplates that many variations of the described nanoemulsions will be useful in the methods of the present invention.
  • three criteria are analyzed. Using the methods and standards described herein, candidate emulsions can be easily tested to determine if they are suitable. First, the desired ingredients are prepared using the methods described herein, to determine if a nanoemulsion can be formed. If a nanoemulsion cannot be formed, the candidate is rejected. Second, the candidate nanoemulsion should form a stable emulsion. A nanoemulsion is stable if it remains in emulsion form for a sufficient period to allow its intended use.
  • the candidate nanoemulsion should have efficacy for its intended use.
  • the emulsions of the invention should kill or disable microorganisms in vitro.
  • the nanoemulsion is exposed to the microorganism for one or more time periods in a side-by-side experiment with an appropriate control sample (e.g., a negative control such as water) and determining if, and to what degree, the nanoemulsion kills or disables the microorganism.
  • an appropriate control sample e.g., a negative control such as water
  • the nanoemulsion of the invention can be provided in many different types of containers and delivery systems.
  • the nanoemulsions are provided as a liquid, lotion, cream or other solid or semi-solid form.
  • the nanoemulsions of the invention may be incorporated into hydrogel formulations.
  • the nanoemulsions can be delivered (e.g., to a subject or customers) in any suitable container. Suitable containers can be used that provide one or more single use or multi-use dosages of the nanoemulsion for the desired application.
  • the nanoemulsions are provided in a suspension or liquid form.
  • Such nanoemulsions can be delivered in any suitable container including spray bottles (e.g., pressurized spray bottles, nebulizers).
  • the present invention is directed to methods of treating and/or preventing a respiratory infection in a subject having Cystic fibrosis (CF).
  • the method comprises administering a nanoemulsion to the subject, wherein the nanoemulsion comprises: (i) water; (ii) at least one organic solvent; (iii) at least one surfactant; and (iv) at least one oil; and wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • the subject is susceptible to or has an infection by one or more bacterial species selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp.
  • the nanoemulsion can be delivered using any pharmaceutically acceptable means, with inhalation being one example of a useful administration method.
  • the invention is directed to a method of treating or preventing an infection in a subject having a burn wound, wherein: (a) the method comprises administering a nanoemulsion to the subject; and (b) the nanoemulsion comprises: (i) water; (ii) at least one organic solvent; (iii) at least one surfactant; and (iv) at least one oil; and wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • the subject is susceptible to or has an infection by one or more gram-negative or gram-positive bacterial species.
  • the bacterial species are selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp.
  • the nanoemulsion can be delivered using any pharmaceutically acceptable means, with inhalation, nebulization, and topical application to mucosal surfaces being examples of useful administration methods.
  • the invention is directed to a method of treating or preventing an Haemophilus influenzae infection in a subject wherein: (a) the method comprises administering a nanoemulsion to the subject having or at risk of having a Haemophilus influenzae infection; (b) the nanoemulsion comprises: (i) water; (ii) at least one organic solvent; (iii) at least one surfactant; and (iv) at least one oil; and (c) wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • the nanoemulsion can be delivered using any pharmaceutically acceptable means, with inhalation, nebulization, and delivery to a mucosal surface being examples of useful administration methods.
  • the nanoemulsion exhibits minimal or no toxicity or side effects.
  • the nanoemulsion does not exhibit resistance to bacteria. This embodiment applies to all methods described herein.
  • the respiratory infection may be associated with a bacterial biofilm, such as a biofilm present in the lungs of a subject.
  • All of the methods of the invention may further comprise administering one or more antibiotics either before, during, or after administration of the nanoemulsion.
  • one or more antibiotics may be incorporated into a nanoemulsion.
  • the nanoemulsion does not exhibit any antagonism with the antibiotic.
  • administration of a nanoemulsion and at least one antibiotic is synergistic as defined by a fractional inhibitory concentration (FIC) index, a fractional bactericidal concentration (FBC) index, or a combination thereof.
  • FEC fractional inhibitory concentration
  • FBC fractional bactericidal concentration
  • antibiotics include, but are not limited to polymyxins (colistin) and aminoglycosides (tobramycin).
  • the methods of the invention may be used to treat or prevent infection by one or more bacterial species selected from the group consisting of Pseudomonas aeruginosa, B. cenocepacia, A. baumannii, Stenotrophomonas maltophilia, Staphylococcus aureus, H influenzae, E. coli, K. pneumoniae , and Proteus mirabilis . All other gram positive or gram negative bacteria are also encompassed by the methods of the invention.
  • the minimum inhibitory concentration (MIC), the minimum bactericidal concentration (MBC), or a combination thereof for the nanoemulsion demonstrate bacteriostatic or bactericidal activity for the nanoemulsion. This embodiment applies to all methods described herein.
  • one or more bacterial species may exhibit resistance against one or more antibiotics.
  • the bacterial species can be methicillin-resistant Staphylococcus aureus (MRSA). This embodiment applies to all methods described herein.
  • the present invention is not limited by the type of subject administered a composition of the present invention.
  • Each of the subjects (e.g., susceptible to respiratory infection) described above may be administered a composition of the present invention.
  • the compositions and methods of the present invention are useful in the treatment of other respiratory diseases and disorders, such as acute bronchitis, bronchiectasis, pneumonia (including ventilator-associated pneumonia, nosocomial pneumonia, viral pneumonia, bacterial pneumonia, mycobacterial pneumonia, fungal pneumonia, eosinophilic pneumonia, and Pneumocystis carinii pneumonia), tuberculosis, cystic fibrosis (CF), emphysema radiation pneumonitis, and respiratory infection associated with inflammation caused by smoking, pulmonary edema, pneumoconiosis, sarcoidiosis, silicosis, asbestosis, berylliosis, coal worker's pneumonoconiosis (CWP), byssinosis, interstitial
  • compositions comprising a nanoemulsion of the present invention may comprise one or more different agents in addition to the nanoemulsion.
  • agents or cofactors include, but are not limited to, adjuvants, surfactants, additives, buffers, solubilizers, chelators, oils, salts, therapeutic agents, drugs, bioactive agents, antibacterials, and antimicrobial agents (e.g., antibiotics, antivirals, etc.).
  • a composition comprising a nanoemulsion of the present invention comprises an agent and/or co-factor that enhance the ability of the nanoemulsion to kill a microbe (e.g., located in the respiratory tract).
  • a microbe e.g., located in the respiratory tract.
  • the presence of one or more co-factors or agents reduces the amount of nanoemulsion required for killing and/or attenuation of growth of a microbe.
  • the present invention is not limited by the type of co-factor or agent used in a therapeutic agent of the present invention.
  • a co-factor or agent used in a nanoemulsion composition is a bioactive agent.
  • the bioactive agent may be a bioactive agent useful in a cell (e.g., a cell expressing a CFTR).
  • Bioactive agents include diagnostic agents such as radioactive labels and fluorescent labels.
  • Bioactive agents also include molecules affecting the metabolism of a cell (e.g., a cell expressing a CFTR), including peptides, nucleic acids, and other natural and synthetic drug molecules.
  • Bioactive agents include, but are not limited to, adrenergic agent; adrenocortical steroid; adrenocortical suppressant; alcohol deterrent; aldosterone antagonist; amino acid; ammonia detoxicant; anabolic; analeptic; analgesic; androgen; anesthesia, adjunct to; anesthetic; anorectic; antagonist; anterior pituitary suppressant; anthelmintic; anti-acne agent; anti-adrenergic; anti-allergic; anti-amebic; anti-androgen; anti-anemic; anti-anginal; anti-anxiety; anti-arthritic; anti-asthmatic; anti-atherosclerotic; antibacterial; anticholelithic; anticholelithogenic; anticholinergic; anticoagulant; anticoccidal; anticonvulsant; antidepressant; antidiabetic; antidiarrheal; antidiuretic; antidote; anti-
  • Molecules useful as antimicrobials can be delivered by the methods and compositions of the invention, such that the respiratory infection is reduced or eliminated.
  • Antibiotics that may find use in co-administration with a composition comprising a nanoemulsion of the present invention include, but are not limited to, agents or drugs that are bactericidal and/or bacteriostatic (e.g., inhibiting replication of bacteria or inhibiting synthesis of bacterial components required for survival of the infecting organism), including, but not limited to, almecillin, amdinocillin, amikacin, amoxicillin, amphomycin, amphotericin B, ampicillin, azacitidine, azaserine, azithromycin, azlocillin, aztreonam, bacampicillin, bacitracin, benzyl penicilloyl-polylysine, bleomycin, candicidin, capreomycin, carbenicillin, cefaclor, cefadroxil, cefamand
  • a composition comprising a nanoemulsion of the present invention comprises one or more mucoadhesives (See, e.g., U.S. Pat. App. No. 20050281843, hereby incorporated by reference in its entirety).
  • the present invention is not limited by the type of mucoadhesive utilized.
  • mucoadhesives are contemplated to be useful in the present invention including, but not limited to, cross-linked derivatives of poly(acrylic acid) (e.g., carbopol and polycarbophil), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides (e.g., alginate and chitosan), hydroxypropyl methylcellulose, lectins, fimbrial proteins, and carboxymethylcellulose.
  • use of a mucoadhesive enhances killing and/or attenuation of growth of a microbe (e.g., exposed to a composition of the present invention) due to an increase in duration and/or amount of exposure to the nanoemulsion that a subject and/or microbe experiences when a mucoadhesive is used compared to the duration and/or amount of exposure to the nanoemulsion in the absence of using the mucoadhesive.
  • a composition of the present invention may comprise sterile aqueous preparations.
  • Acceptable vehicles and solvents include, but are not limited to, water, Ringer's solution, phosphate buffered saline and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed mineral or non-mineral oil may be employed including synthetic mono-ordi-glycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Carrier formulations suitable for mucosal, pulmonary, subcutaneous, intramuscular, intraperitoneal, intravenous, or administration via other routes may be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • a composition comprising a nanoemulsion of the present invention can be used therapeutically (e.g., to kill and/or attenuate growth of an existing infection) or as a prophylactic (e.g., to prevent microbial growth and/or colonization (e.g., to prevent signs or symptoms of disease)).
  • a composition comprising a nanoemulsion of the present invention can be administered to a subject via a number of different delivery routes and methods.
  • compositions of the present invention can be administered to a subject (e.g., mucosally or by pulmonary route) by multiple methods, including, but not limited to: being suspended in a solution and applied to a surface; being suspended in a solution and sprayed onto a surface using a spray applicator; being mixed with a mucoadhesive and applied (e.g., sprayed or wiped) onto a surface (e.g., mucosal or pulmonary surface); being placed on or impregnated onto a nasal and/or pulmonary applicator and applied; being applied by a controlled-release mechanism; applied using a nebulizer, aerosolized, being applied as a liposome; or being applied on a polymer.
  • a subject e.g., mucosally or by pulmonary route
  • multiple methods including, but not limited to: being suspended in a solution and applied to a surface; being suspended in a solution and sprayed onto a surface using a spray applicator; being mixed
  • compositions of the present invention are administered mucosally (e.g., using standard techniques; See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995 (e.g., for mucosal delivery techniques, including intranasal and pulmonary techniques), as well as European Publication No. 517,565 and Illum et al., J. Controlled Rel., 1994, 29:133-141 (e.g., for techniques of intranasal administration), each of which is hereby incorporated by reference in its entirety).
  • the present invention is not limited by the route of administration.
  • compositions of the present invention may also be administered via the oral route.
  • a composition comprising a nanoemulsion may comprise a pharmaceutically acceptable excipient and/or include alkaline buffers, or enteric capsules.
  • Formulations for nasal delivery may include those with dextran or cyclodextran and saponin as an adjuvant.
  • a nanoemulsion of the present invention is administered via a pulmonary delivery route and/or means.
  • an aqueous solution containing the nanoemulsion is gently and thoroughly mixed to form a solution.
  • the solution is sterile filtered (e.g., through a 0.2 micron filter) into a sterile, enclosed vessel. Under sterile conditions, the solution is passed through an appropriately small orifice to make droplets (e.g., between 0.1 and 10 microns).
  • the particles may be administered using any of a number of different applicators. Suitable methods for manufacture and administration are described in the following U.S. Pat. Nos. 6,592,904; 6,518,239; 6,423,344; 6,294,204; 6,051,256 and 5,997,848 to INHALE (now NEKTAR); and U.S. Pat. No. 5,985,309; RE37,053; U.S. Pat. Nos. 6,436,443; 6,447,753; 6,503,480; and 6,635,283, to Edwards, et al. (MIT, AIR), each of which is hereby incorporated
  • compositions of the present invention are administered by pulmonary delivery.
  • a composition of the present invention can be delivered to the lungs of a subject (e.g., a human) via inhalation (See, e.g., Adjei, et al. Pharmaceutical Research 1990; 7:565-569; Adjei, et al. Int. J. Pharmaceutics 1990; 63:135-144; Braquet, et al. J. Cardiovascular Pharmacology 1989 143-146; Hubbard, et al. (1989) Annals of Internal Medicine, Vol. III, pp. 206-212; Smith, et al. J. Clin. Invest.
  • a composition comprising a nanoemulsion is administered to a subject by more than one route or means (e.g., administered via pulmonary route as well as a mucosal route).
  • nebulizers metered dose inhalers
  • powder inhalers all of which are familiar to those skilled in the art.
  • Some specific examples of commercially available devices suitable for the practice of this invention are the ULTRAVENT nebulizer (Mallinckrodt Inc., St. Louis, Mo.); the ACORN II nebulizer (Marquest Medical Products, Englewood, Colo.); the VENTOLIN metered dose inhaler (Glaxo Inc., Research Triangle Park, N.C.); and the SPINHALER powder inhaler (Fisons Corp., Bedford, Mass.).
  • each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants, surfactants, carriers and/or other agents useful in therapy. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated.
  • a composition comprising a nanoemulsion of the present invention may be used to protect and/or treat a subject susceptible to, or suffering from, a disease by means of administering compositions comprising a nanoemulsion by mucosal, intramuscular, intraperitoneal, intradermal, transdermal, pulmonary, intravenous, subcutaneous or other route of administration described herein.
  • Methods of systemic administration of the nanoemulsion and/or agent co-administered with the nanoemulsion may include conventional syringes and needles, or devices designed for ballistic delivery (See, e.g., WO 99/27961, hereby incorporated by reference), or needleless pressure liquid jet device (See, e.g., U.S.
  • the present invention provides a delivery device for systemic administration, pre-filled with the nanoemulsion composition of the present invention.
  • the present invention is not limited by the type of subject administered a composition of the present invention. Indeed, a wide variety of subjects are contemplated to be benefited from administration of a composition of the present invention.
  • the subject is a human.
  • human subjects are of any age (e.g., adults, children, infants, etc.) that have been or are likely to become exposed to a microorganism.
  • the human subjects are subjects that are more likely to receive a direct exposure to pathogenic microorganisms or that are more likely to display signs and symptoms of disease after exposure to a pathogen (e.g., subjects with CF or asthma, subjects in the armed forces, government employees, frequent travelers, persons attending or working in a school or daycare, health care workers, an elderly person, an immunocompromised person, and emergency service employees (e.g., police, fire, EMT employees)).
  • a pathogen e.g., subjects with CF or asthma, subjects in the armed forces, government employees, frequent travelers, persons attending or working in a school or daycare, health care workers, an elderly person, an immunocompromised person, and emergency service employees (e.g., police, fire, EMT employees)
  • any one or all members of the general public can be administered a composition of the present invention (e.g., to prevent the occurrence or spread of disease).
  • compositions and methods of the present invention are utilized to treat a group of people (e.g., a population of a region, city, state and/or country) for their own health (e.g., to prevent or treat disease) and/or to prevent or reduce the risk of disease spread from animals (e.g., birds, cattle, sheep, pigs, etc.) to humans.
  • animals e.g., birds, cattle, sheep, pigs, etc.
  • the subjects are non-human mammals (e.g., pigs, cattle, goats, horses, sheep, or other livestock; or mice, rats, rabbits or other animal).
  • compositions and methods of the present invention are utilized in research settings (e.g., with research animals).
  • a composition comprising a nanoemulsion of the present invention can be administered (e.g., to a subject (e.g., via pulmonary and/or mucosal route) or to microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria (e.g., residing on or within the respiratory system of a subject and/or on or within a burn wound))) as a therapeutic or as a prophylactic to prevent microbial infection.
  • a subject e.g., via pulmonary and/or mucosal route
  • microbes e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria (e.g., residing on or within the respiratory system of a subject and/or on or within a burn wound))
  • bacteria e.g., opportunistic and/or pathogenic bacteria (e.g., residing on or within the respiratory system of a subject and/or on or within a burn
  • the present invention provides a method of altering microbial (e.g., bacterial (e.g., opportunistic and/or pathogenic bacterial) growth comprising administering a composition comprising a nanoemulsion to the microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria).
  • a composition comprising a nanoemulsion to the microbes e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) kills the microbes.
  • a composition comprising nanoemulsion to the microbes inhibits growth of the microbes.
  • a composition comprising a nanoemulsion can be administered to microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria (e.g., residing within the respiratory tract)) via a number of delivery routes and/or mechanisms.
  • compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions.
  • the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • additional materials useful in physically formulating various dosage forms of the compositions of the present invention such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • such materials when added, preferably do not unduly interfere with the biological activities of the components of the compositions of the present invention.
  • nanoemulsion compositions of the present invention are administered in the form of a pharmaceutically acceptable salt.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include, but are not limited to, acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v).
  • Suitable preservatives may include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • a composition comprising a nanoemulsion is co-administered with one or more antibiotics.
  • one or more antibiotics may be administered with, before and/or after administration of a composition comprising a nanoemulsion.
  • the present invention is not limited by the type of antibiotic co-administered.
  • antibiotics may be co-administered including, but not limited to, ⁇ -lactam antibiotics, penicillins (such as natural penicillins, aminopenicillins, penicillinase-resistant penicillins, carboxy penicillins, ureido penicillins), cephalosporins (first generation, second generation, and third generation cephalosporins), and other ⁇ -lactams (such as imipenem, monobactams), ⁇ -lactamase inhibitors, vancomycin, aminoglycosides and spectinomycin, tetracyclines, chloramphenicol, erythromycin, lincomycin, clindamycin, rifampin, metronidazole, polymyxins, doxycycline, quinolones (e.g., ciprofloxacin), sulfonamides, trimethoprim, and quinolines.
  • penicillins such as natural penicillins, aminopenicillins, penicillinas
  • these agents include agents that inhibit cell wall synthesis (e.g., penicillins, cephalosporins, cycloserine, vancomycin, bacitracin); and the imidazole antifungal agents (e.g., miconazole, ketoconazole and clotrimazole); agents that act directly to disrupt the cell membrane of the microorganism (e.g., detergents such as polmyxin and colistimethate and the antifungals nystatin and amphotericin B); agents that affect the ribosomal subunits to inhibit protein synthesis (e.g., chloramphenicol, the tetracyclines, erthromycin and clindamycin); agents that alter protein synthesis and lead to cell death (e.g., aminoglycosides); agents that affect nucleic acid metabolism (e.g., the rifamycins and the quinolones); the antimetabolites (e.g., trimethoprim and sulf
  • the present invention also includes methods involving co-administration of a composition comprising a nanoemulsion with one or more additional active and/or anti-infective agents.
  • the agents may be administered concurrently or sequentially.
  • the compositions described herein are administered prior to the other active agent(s).
  • the pharmaceutical formulations and modes of administration may be any of those described herein.
  • the two or more co-administered agents may each be administered using different modes (e.g., routes) or different formulations.
  • the additional agents to be co-administered e.g., antibiotics, a second type of nanoemulsion, etc.
  • a composition comprising a nanoemulsion is administered to a subject via more than one route.
  • a subject may benefit from receiving mucosal administration (e.g., nasal administration or other mucosal routes described herein) and, additionally, receiving one or more other routes of administration (e.g., pulmonary administration (e.g., via a nebulizer, inhaler, or other methods described herein.
  • mucosal administration e.g., nasal administration or other mucosal routes described herein
  • one or more other routes of administration e.g., pulmonary administration (e.g., via a nebulizer, inhaler, or other methods described herein.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the compositions, increasing convenience to the subject and a physician.
  • release delivery systems are available and known to those of ordinary skill in the art. They include polymer based systems such as poly (lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.
  • Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109, hereby incorporated by reference.
  • Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • Specific examples include, but are not limited to: (a) erosional systems in which an agent of the invention is contained in a form within a matrix such as those described in U.S. Pat. Nos.
  • a composition comprising a nanoemulsion of the present invention comprises a suitable amount of the nanoemulsion to kill and/or attenuate growth of microbes (e.g., pathogenic microbes (e.g., pathogenic bacteria, viruses, etc.)) in a subject when administered to the subject.
  • microbes e.g., pathogenic microbes (e.g., pathogenic bacteria, viruses, etc.)
  • the present invention is not limited by the amount of nanoemulsion used.
  • the amount of nanoemulsion in a composition comprising a nanoemulsion is selected as that amount which kills and/or attenuates microbial growth without significant, adverse side effects.
  • the amount will vary depending upon which specific nanoemulsion(s) is/are employed, and can vary from subject to subject, depending on a number of factors including, but not limited to, the species, age and general condition (e.g., health) of the subject, and the mode of administration.
  • Procedures for determining the appropriate amount of nanoemulsion administered to a subject to kill and/or attenuate growth of a microbe (e.g., pathogenic microbe (e.g., pathogenic bacteria, viruses, etc.)) in a subject can be readily determined using known means by one of ordinary skill in the art.
  • each dose e.g., of a composition comprising a nanoemulsion (e.g., administered to a subject to kill and/or attenuate microbial growth)
  • An optimal amount for a particular administration e.g., to kill and/or attenuate microbial growth
  • each dose e.g., of a composition comprising a nanoemulsion (e.g., administered to a subject to kill and/or attenuate microbial growth)
  • a composition comprising a nanoemulsion e.g., administered to a subject to kill and/or attenuate microbial growth
  • each dose is from 0.001 to 40% or more (e.g., 0.001-10%, 0.5-5%, 1-3%, 2%, 6%, 10%, 15%, 20%, 30%, 40% or more) by weight nanoemulsion.
  • the present invention is not limited by the duration of time a nanoemulsion is administered to a subject (e.g., to kill and/or attenuate microbial growth).
  • a nanoemulsion is administered one or more times (e.g. twice, three times, four times or more) daily.
  • a composition comprising a nanoemulsion is administered one or more times a day until an infection is eradicated or microbial growth and/or presence has been reduced to a desired level.
  • a composition comprising a nanoemulsion of the present invention is formulated in a concentrated dose that can be diluted prior to administration to a subject.
  • dilutions of a concentrated composition may be administered to a subject such that the subject receives any one or more of the specific dosages provided herein.
  • dilution of a concentrated composition may be made such that a subject is administered (e.g., in a single dose) a composition comprising 0.5-50% of the nanoemulsion present in the concentrated composition.
  • Concentrated compositions are contemplated to be useful in a setting in which large numbers of subjects may be administered a composition of the present invention (e.g., a hospital).
  • a composition comprising a nanoemulsion of the present invention is stable at room temperature for more than 1 week, in some embodiments for more than 2 weeks, in some embodiments for more than 3 weeks, in some embodiments for more than 4 weeks, in some embodiments for more than 5 weeks, and in some embodiments for more than 6 weeks.
  • Dosage units may be proportionately increased or decreased based on several factors including, but not limited to, the weight, age, and health status of the subject. In addition, dosage units may be increased or decreased for subsequent administrations.
  • a composition comprising a nanoemulsion is administered to a subject under conditions such that microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria)) are killed.
  • a composition comprising a nanoemulsion is administered to a subject under conditions such that microbial (e.g., bacterial (e.g., opportunistic and/or pathogenic bacterial) growth is prohibited and/or attenuated.
  • microbial e.g., bacterial (e.g., opportunistic and/or pathogenic bacterial) growth is prohibited and/or attenuated.
  • greater than 90% e.g., greater than 95%, 98%, 99%, all detectable
  • microbes e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) are killed.
  • microbe e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) presence.
  • reduction and/or killing is observed in one hour or less (e.g., 45 minutes, 30 minutes, 15 minutes, or less).
  • reduction and/or killing is observed in 6 hours or less (e.g., 5 hours, 4, hours, 3 hours, two hours or less than one hour).
  • reduction and/or killing is observed in two days or less following initial treatment (e.g., less than 24 hours, less than 20 hours, 18 hours or less).
  • the reduction and/or killing is observed in three days or less, four days or less, or five days or less.
  • a composition comprising a nanoemulsion of the present invention finds use where the nature of the infectious and/or disease causing agent (e.g., causing signs, symptoms or indications of respiratory infection) is known, as well as where the nature of the infectious and/or disease causing agent is unknown (e.g., in emerging disease (e.g., of pandemic proportion (e.g., influenza or other outbreaks of disease))).
  • the present invention contemplates use of the compositions of the present invention in treatment of or prevention of infections associated with an emergent infectious and/or disease causing agent yet to be identified (e.g., isolated and/or cultured from a diseased person but without genetic, biochemical or other characterization of the infectious and/or disease causing agent).
  • compositions and methods of the present invention will find use in various settings, including research settings.
  • compositions and methods of the present invention also find use in studies of the immune system (e.g., characterization of adaptive immune responses (e.g., protective immune responses (e.g., mucosal or systemic immunity))).
  • Uses of the compositions and methods provided by the present invention encompass human and non-human subjects and samples from those subjects, and also encompass research applications using these subjects.
  • Compositions and methods of the present invention are also useful in studying and optimizing nanoemulsions, immunogens, and other components and for screening for new components. Thus, it is not intended that the present invention be limited to any particular subject and/or application setting.
  • compositions of the present invention are useful for preventing and/or treating a wide variety of diseases and infections caused by viruses, bacteria, parasites, and fungi.
  • the compositions can also be used in order to prepare antibodies, both polyclonal and monoclonal (e.g., for diagnostic purposes), as well as for immunopurification of an antigen of interest.
  • the present invention provides a kit comprising a composition comprising a nanoemulsion.
  • the kit further provides a device for administering the composition.
  • the present invention is not limited by the type of device included in the kit.
  • the device is configured for pulmonary application of the composition of the present invention (e.g., a nasal inhaler or nasal mister).
  • a kit comprises a composition comprising a nanoemulsion in a concentrated form (e.g., that can be diluted prior to administration to a subject).
  • kits are present within a single container (e.g., vial or tube).
  • each kit component is located in a single container (e.g., vial or tube).
  • one or more kit components are located in a single container (e.g., vial or tube) with other components of the same kit being located in a separate container (e.g., vial or tube).
  • a kit comprises a buffer.
  • the kit further comprises instructions for use.
  • the present invention provides immunogenic nanoemulsion compositions and methods of using the same for the induction of immune responses (e.g., innate and/or adaptive immune responses (e.g., for generation of host immunity against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans , etc.))).
  • immune responses e.g., innate and/or adaptive immune responses (e.g., for generation of host immunity against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans , etc.)
  • Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine (e.g., vaccination)) and research applications.
  • an immunogenic composition e.g., comprising a nanoemulsion and Bukholderia antigen
  • induces e.g., when administered to a subject
  • both systemic and mucosal immunity e.g., when administered to a subject
  • administration of a composition comprising a nanoemulsion and Burkholderia antigen to a subject results in protection against an exposure (e.g., a mucosal and/or respiratory exposure) to Burkholderia .
  • mucosal administration e.g., vaccination
  • Burkholderia e.g., Burkholderia cepacia
  • pathogens that invade at mucosal surfaces
  • the present invention provides compositions and methods for stimulating mucosal immunity (e.g., a protective IgA response) towards a pathogen (e.g., pathogenic species of Burkholderia, Haemophilus, Staphylococcus or other bacterial genus) in a subject.
  • a pathogen e.g., pathogenic species of Burkholderia, Haemophilus, Staphylococcus or other bacterial genus
  • the present invention provides a composition (e.g., a composition comprising a nanoemulsion and Burkholderia (e.g., Burkholderia cenocepacia ) antigen (e.g., an immunogenic polypeptide comprising an amino acid sequence of SEQ ID NO. 1 or SEQ ID NOS. 2-16)) to serve as a mucosal vaccine.
  • a composition e.g., a composition comprising a nanoemulsion and Burkholderia (e.g., Burkholderia cenocepacia ) antigen (e.g., an immunogenic polypeptide comprising an amino acid sequence of SEQ ID NO. 1 or SEQ ID NOS. 2-16)
  • Burkholderia e.g., Burkholderia cenocepacia
  • an immunogenic polypeptide comprising an amino acid sequence of SEQ ID NO. 1 or SEQ ID NOS. 2-16
  • an immunogenic composition comprising nanoemulsion (NE) and Burkholderia antigen is produced with NE and killed whole cell bacteria of the genus Burkholderia (e.g., Burkholderia cepacia (e.g., killed using nanoemulsion, alcohol (e.g., ethanol), or other methods), isolated, purified and/or recombinant protein and/or saccharide component of Burkholderia (e.g., protein/peptide (e.g., Burkholderia -derived protein, live-virus-vector-derived protein, recombinant protein, recombinant denatured protein/antigens, small peptide segments protein/antigen).
  • Burkholderia e.g., Burkholderia cepacia (e.g., killed using nanoemulsion, alcohol (e.g., ethanol), or other methods
  • isolated, purified and/or recombinant protein and/or saccharide component of Burkholderia e
  • the present invention provides a composition for generating an immune response comprising a NE and an immunogen (e.g., a purified, isolated or synthetic Burkholderia protein or derivative, variant, or analogue thereof; or, one or more species of Burkholderia (e.g., Burkholderia multivorans (e.g., killed and or inactivated whole cell bacteria).
  • an immunogen e.g., a purified, isolated or synthetic Burkholderia protein or derivative, variant, or analogue thereof; or, one or more species of Burkholderia (e.g., Burkholderia multivorans (e.g., killed and or inactivated whole cell bacteria).
  • an immunogen e.g., a purified, isolated or synthetic Burkholderia protein or derivative, variant, or analogue thereof; or, one or more species of Burkholderia (e.g., Burkholderia multivorans (e.g., killed and or inactivated whole cell bacteria).
  • generation of an immune response (e.g., resulting from administration of a composition comprising a nanoemulsion and an immunogen) provides total or partial immunity to the subject (e.g., from signs, symptoms or conditions of a disease associated with Burkholderia infection (e.g., strep throat, meningitis, bacterial pneumoniae , endocarditis, erysipelas and/or necrotizing fasciitis)).
  • Burkholderia infection e.g., strep throat, meningitis, bacterial pneumoniae , endocarditis, erysipelas and/or necrotizing fasciitis
  • protection and/or immunity from disease is due to adaptive (e.g., acquired) immune responses (e.g., immune responses mediated by B and T cells following exposure to a NE comprising an immunogen of the present invention (e.g., immune responses that exhibit increased specificity and reactivity towards Burkholderia (e.g., Burkholderia cepacia )).
  • adaptive immune responses e.g., immune responses mediated by B and T cells following exposure to a NE comprising an immunogen of the present invention (e.g., immune responses that exhibit increased specificity and reactivity towards Burkholderia (e.g., Burkholderia cepacia )).
  • compositions and methods of the present invention are used prophylactically or therapeutically to prevent or attenuate a sign, symptom or condition associated with Burkholderia (e.g., Burkholderia cepacia )).
  • Burkholderia e.g., Burkholderia cepacia
  • a NE comprising an immunogen e.g., a Burkholderia (e.g., Burkholderia cepacia ) antigen) is administered alone.
  • a composition comprising a NE and an immunogen e.g., a Burkholderia (e.g., Burkholderia cepacia ) antigen) comprises one or more other agents (e.g., a pharmaceutically acceptable carrier, adjuvant, excipient, and the like).
  • a composition for stimulating an immune response of the present invention is administered in a manner to induce a humoral immune response.
  • a composition for stimulating an immune response of the present invention is administered in a manner to induce a cellular (e.g., cytotoxic T lymphocyte) immune response, rather than a humoral response.
  • a composition comprising a NE and an immunogen of the present invention induces both a cellular and humoral immune response.
  • the present invention is not limited by the isotype, strain or species of Burkholderia (e.g., Burkholderia cepacia ) used in a composition comprising a NE and immunogen.
  • Burkholderia e.g., Burkholderia cepacia
  • each Burkholderia e.g., Burkholderia cepacia family member alone, or in combination with another family member, may be used to generate a composition comprising a NE and an immunogen (e.g., used to generate an immune response) of the present invention.
  • an immunogen e.g., used to generate an immune response
  • Exemplary species of Burkholderia are described herein.
  • the Burkholderia (e.g., Burkholderia cepacia ) species utilized is a modified (e.g., genetically modified (e.g., naturally modified via natural selection or modified using recombinant genetic techniques)) species that displays greater pathogenic capacity (e.g., causes more sever Burkholderia —(e.g., Burkholderia cepacia )-induced disease (e.g., comprising enhanced and/or more severe respiratory infection, etc.)).
  • any one or more members of the genus Burkholderia is utilized in an immunoreactive composition of the invention including but not limited to B. cenocepacia, B. dolosa, B. multivorans, B.
  • the present invention is not limited by the Burkholderia strain used. Indeed, a variety of Burkholderia strains are contemplated to be useful in the present invention including, but not limited to, classical strains, attenuated strains, non-replicating strains, modified strains (e.g., genetically or mechanically modified strains (e.g., to become more or less virulent)), or other serially diluted strains of Burkholderia .
  • a composition comprising a NE and immunogen may comprise one or more strains of Burkholderia cenocepacia and/or other type of Burkholderia (e.g., Burkholderia multivorans). Additionally, a composition comprising a NE and immunogen may comprise one or more strains of Burkholderia and, in addition, one or more strains of a non- Burkholderia immunogen.
  • the immunogen may comprise one or more antigens derived from a pathogen (e.g., Burkholderia ).
  • a pathogen e.g., Burkholderia
  • the immunogen is a purified, recombinant, synthetic, or otherwise isolated protein (e.g., added to the NE to generate an immunogenic composition).
  • the immunogenic protein may be a derivative, analogue or otherwise modified (e.g., conjugated) form of a protein from a pathogen.
  • compositions comprising a NE and immunogen of the present invention may comprise one or more different agents in addition to the NE and immunogen.
  • agents or cofactors include, but are not limited to, adjuvants, surfactants, additives, buffers, solubilizers, chelators, oils, salts, therapeutic agents, drugs, bioactive agents, antibacterials, and antimicrobial agents (e.g., antibiotics, antivirals, etc.).
  • a composition comprising a NE and immunogen of the present invention comprises an agent and/or co-factor that enhance the ability of the immunogen to induce an immune response (e.g., an adjuvant).
  • an agent and/or co-factor that enhance the ability of the immunogen to induce an immune response e.g., an adjuvant.
  • the presence of one or more co-factors or agents reduces the amount of immunogen required for induction of an immune response (e.g., a protective immune response (e.g., protective immunization)).
  • the presence of one or more co-factors or agents can be used to skew the immune response towards a cellular (e.g., T cell mediated) or humoral (e.g., antibody mediated) immune response.
  • the present invention is not limited by the type of co-factor or agent used in a therapeutic agent of the present invention.
  • Adjuvants are described in general in Vaccine Design—the Subunit and Adjuvant Approach, edited by Powell and Newman, Plenum Press, New York, 1995.
  • the present invention is not limited by the type of adjuvant utilized (e.g., for use in a composition (e.g., pharmaceutical composition) comprising a NE and immunogen).
  • suitable adjuvants include an aluminum salt such as aluminum hydroxide gel (alum) or aluminum phosphate.
  • an adjuvant may be a salt of calcium, iron or zinc, or may be an insoluble suspension of acylated tyrosine, or acylated sugars, cationically or anionically derivatised polysaccharides, or polyphosphazenes.
  • compositions comprising a NE and immunogen of the present invention comprises one or more adjuvants that induce a Th1-type response.
  • a composition comprising a NE and immunogen of the present invention comprises one or more adjuvants that induce a Th2-type response.
  • an immune response is generated to an antigen through the interaction of the antigen with the cells of the immune system.
  • Immune responses may be broadly categorized into two categories: humoral and cell mediated immune responses (e.g., traditionally characterized by antibody and cellular effector mechanisms of protection, respectively). These categories of response have been termed Th1-type responses (cell-mediated response), and Th2-type immune responses (humoral response).
  • Stimulation of an immune response can result from a direct or indirect response of a cell or component of the immune system to an intervention (e.g., exposure to an immunogen).
  • Immune responses can be measured in many ways including activation, proliferation or differentiation of cells of the immune system (e.g., B cells, T cells, dendritic cells, APCs, macrophages, NK cells, NKT cells etc.); up-regulated or down-regulated expression of markers and cytokines; stimulation of IgA, IgM, or IgG titer; splenomegaly (including increased spleen cellularity); hyperplasia and mixed cellular infiltrates in various organs.
  • Other responses, cells, and components of the immune system that can be assessed with respect to immune stimulation are known in the art.
  • compositions and methods of the present invention induce expression and secretion of cytokines (e.g., by macrophages, dendritic cells and CD4+ T cells). Modulation of expression of a particular cytokine can occur locally or systemically. It is known that cytokine profiles can determine T cell regulatory and effector functions in immune responses.
  • Th1-type cytokines can be induced, and thus, the immunostimulatory compositions of the present invention can promote a Th1 type antigen-specific immune response including cytotoxic T-cells (e.g., thereby avoiding unwanted Th2 type immune responses (e.g., generation of Th2 type cytokines (e.g., IL-13) involved in enhancing the severity of disease (e.g., IL-13 induction of mucus formation))).
  • Th2 type cytokines e.g., IL-13
  • Cytokines play a role in directing the T cell response.
  • Helper (CD4+) T cells orchestrate the immune response of mammals through production of soluble factors that act on other immune system cells, including B and other T cells. Most mature CD4+ T helper cells express one of two cytokine profiles: Th1 or Th2.
  • Th1-type CD4+ T cells secrete IL-2, IL-3, IFN- ⁇ , GM-CSF and high levels of TNF- ⁇ .
  • Th2 cells express IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, GM-CSF and low levels of TNF- ⁇ .
  • Th1 type cytokines promote both cell-mediated immunity, and humoral immunity that is characterized by immunoglobulin class switching to IgG2a in mice and IgG1 in humans. Th1 responses may also be associated with delayed-type hypersensitivity and autoimmune disease. Th2 type cytokines induce primarily humoral immunity and induce class switching to IgG1 and IgE.
  • the antibody isotypes associated with Th1 responses generally have neutralizing and opsonizing capabilities whereas those associated with Th2 responses are associated more with allergic responses.
  • IL-12 and IFN- ⁇ are positive Th1 and negative Th2 regulators.
  • IL-12 promotes IFN- ⁇ production, and IFN- ⁇ provides positive feedback for IL-12.
  • IL-4 and IL-10 appear important for the establishment of the Th2 cytokine profile and to down-regulate Th1 cytokine production.
  • the present invention provides a method of stimulating a Th1-type immune response in a subject comprising administering to a subject a composition comprising a NE and an immunogen.
  • the present invention provides a method of stimulating a Th2-type immune response in a subject (e.g., if balancing of a T cell mediated response is desired) comprising administering to a subject a composition comprising a NE and an immunogen.
  • adjuvants can be used (e.g., can be co-administered with a composition of the present invention) to skew an immune response toward either a Th1 or Th2 type immune response.
  • adjuvants that induce Th2 or weak Th1 responses include, but are not limited to, alum, saponins, and SB-As4.
  • adjuvants that induce Th1 responses include but are not limited to MPL, MDP, ISCOMS, IL-12, IFN- ⁇ , and SB-AS2.
  • Th1-type immunogens can be used (e.g., as an adjuvant) in compositions and methods of the present invention. These include, but are not limited to, the following.
  • monophosphoryl lipid A e.g., in particular 3-de-O-acylated monophosphoryl lipid A (3D-MPL)
  • 3D-MPL is a well known adjuvant manufactured by Ribi Immunochem, Montana. Chemically it is often supplied as a mixture of 3-de-O-acylated monophosphoryl lipid A with either 4, 5, or 6 acylated chains.
  • diphosphoryl lipid A, and 3-O-deacylated variants thereof are used.
  • 3D-MPL is used in the form of a particulate formulation (e.g., having a small particle size less than 0.2 ⁇ m in diameter, described in EP 0 689 454, hereby incorporated by reference in its entirety).
  • saponins are used as an immunogen (e.g., Th1-type adjuvant) in a composition of the present invention.
  • Saponins are well known adjuvants (See, e.g., Lacaille-Dubois and Wagner (1996) Phytomedicine vol 2 pp 363-386).
  • Examples of saponins include Quil A (derived from the bark of the South American tree Quillaja Saponaria Molina), and fractions thereof (See, e.g., U.S. Pat. No. 5,057,540; Kensil, Crit. Rev Ther Drug Carrier Syst, 1996, 12 (1-2):1-55; and EP 0 362 279, each of which is hereby incorporated by reference in its entirety).
  • haemolytic saponins QS7, QS17, and QS21 HPLC purified fractions of Quil A; See, e.g., Kensil et al. (1991). J. Immunology 146, 431-437, U.S. Pat. No. 5,057,540; WO 96/33739; WO 96/11711 and EP 0 362 279, each of which is hereby incorporated by reference in its entirety).
  • QS21 and polysorbate or cyclodextrin See, e.g., WO 99/10008, hereby incorporated by reference in its entirety.
  • an immunogenic oligonucleotide containing unmethylated CpG dinucleotides (“CpG”) is used as an adjuvant in the present invention.
  • CpG is an abbreviation for cytosine-guanosine dinucleotide motifs present in DNA.
  • CpG is known in the art as being an adjuvant when administered by both systemic and mucosal routes (See, e.g., WO 96/02555, EP 468520, Davis et al., J. Immunol, 1998, 160(2):870-876; McCluskie and Davis, J. Immunol., 1998, 161(9):4463-6; and U.S. Pat. App. No.
  • the immunostimulatory sequence is Purine-Purine-C-G-pyrimidine-pyrimidine; wherein the CG motif is not methylated.
  • CpG oligonucleotides activate various immune subsets including natural killer cells (which produce IFN- ⁇ ) and macrophages.
  • CpG oligonucleotides are formulated into a composition of the present invention for inducing an immune response.
  • a free solution of CpG is co-administered together with an antigen (e.g., present within a NE solution (See, e.g., WO 96/02555; hereby incorporated by reference).
  • a CpG oligonucleotide is covalently conjugated to an antigen (See, e.g., WO 98/16247, hereby incorporated by reference), or formulated with a carrier such as aluminium hydroxide (See, e.g., Brazolot-Millan et al., Proc. Natl. Acad Sci., USA, 1998, 95(26), 15553-8).
  • adjuvants such as Complete Freunds Adjuvant and Incomplete Freunds Adjuvant, cytokines (e.g., interleukins (e.g., IL-2, IFN- ⁇ , IL-4, etc.), macrophage colony stimulating factor, tumor necrosis factor, etc.), detoxified mutants of a bacterial ADP-ribosylating toxin such as a cholera toxin (CT), a pertussis toxin (PT), or an E.
  • cytokines e.g., interleukins (e.g., IL-2, IFN- ⁇ , IL-4, etc.)
  • macrophage colony stimulating factor e.g., tumor necrosis factor, etc.
  • a bacterial ADP-ribosylating toxin such as a cholera toxin (CT), a pertussis toxin (PT), or an E.
  • CT cholera toxin
  • PT pertussis toxin
  • Coli heat-labile toxin particularly LT-K63 (where lysine is substituted for the wild-type amino acid at position 63)
  • LT-R72 where arginine is substituted for the wild-type amino acid at position 72
  • CT-S109 where serine is substituted for the wild-type amino acid at position 109
  • PT-K9/G129 where lysine is substituted for the wild-type amino acid at position 9 and glycine substituted at position 129)
  • adjuvants that find use in the present invention include poly(di(carboxylatophenoxy)phosphazene (PCPP polymer; Virus Research Institute, USA); derivatives of lipopolysaccharides such as monophosphoryl lipid A (MPL; Ribi ImmunoChem Research, Inc., Hamilton, Mont.), muramyl dipeptide (MDP; Ribi) and threonyl-muramyl dipeptide (t-MDP; Ribi); OM-174 (a glucosamine disaccharide related to lipid A; OM Pharma SA, Meyrin, Switzerland); and Leishmania elongation factor (a purified Leishmania protein; Corixa Corporation, Seattle, Wash.).
  • PCPP polymer polymer
  • Virus Research Institute, USA poly(di(carboxylatophenoxy)phosphazene
  • MPL monophosphoryl lipid A
  • MDP muramyl dipeptide
  • t-MDP threonyl-muramyl
  • Adjuvants may be added to a composition comprising a NE and an immunogen, or, the adjuvant may be formulated with carriers, for example liposomes, or metallic salts (e.g., aluminium salts (e.g., aluminium hydroxide)) prior to combining with or co-administration with a composition comprising a NE and an immunogen.
  • carriers for example liposomes, or metallic salts (e.g., aluminium salts (e.g., aluminium hydroxide)) prior to combining with or co-administration with a composition comprising a NE and an immunogen.
  • a composition comprising a NE and an immunogen comprises a single adjuvant. In other embodiments, a composition comprising a NE and an immunogen comprises two or more adjuvants (See, e.g., WO 94/00153; WO 95/17210; WO 96/33739; WO 98/56414; WO 99/12565; WO 99/11241; and WO 94/00153, each of which is hereby incorporated by reference in its entirety).
  • a composition comprising a NE and an immunogen of the present invention comprises one or more mucoadhesives (See, e.g., U.S. Pat. App. No. 20050281843, hereby incorporated by reference in its entirety).
  • the present invention is not limited by the type of mucoadhesive utilized.
  • mucoadhesives are contemplated to be useful in the present invention including, but not limited to, cross-linked derivatives of poly(acrylic acid) (e.g., carbopol and polycarbophil), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides (e.g., alginate and chitosan), hydroxypropyl methylcellulose, lectins, fimbrial proteins, and carboxymethylcellulose.
  • a mucoadhesive e.g., in a composition comprising a NE and immunogen
  • a mucoadhesive enhances induction of an immune response in a subject (e.g., administered a composition of the present invention) due to an increase in duration and/or amount of exposure to an immunogen that a subject experiences when a mucoadhesive is used compared to the duration and/or amount of exposure to an immunogen in the absence of using the mucoadhesive.
  • a composition of the present invention may comprise sterile aqueous preparations.
  • Acceptable vehicles and solvents include, but are not limited to, water, Ringer's solution, phosphate buffered saline and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed mineral or non-mineral oil may be employed including synthetic mono-ordi-glycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Carrier formulations suitable for mucosal, subcutaneous, intramuscular, intraperitoneal, intravenous, or administration via other routes may be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • a composition comprising a NE and an immunogen of the present invention can be used therapeutically (e.g., to enhance an immune response) or as a prophylactic (e.g., for immunization (e.g., to prevent signs or symptoms of disease)).
  • a composition comprising a NE and an immunogen of the present invention can be administered to a subject via a number of different delivery routes and methods.
  • an immunogenic compositions of the invention can be administered to a subject (e.g., mucosally (e.g., nasal mucosa, vaginal mucosa, etc.)) by multiple methods, including, but not limited to: being suspended in a solution and applied to a surface; being suspended in a solution and sprayed onto a surface using a spray applicator; being mixed with a mucoadhesive and applied (e.g., sprayed or wiped) onto a surface (e.g., mucosal surface); being placed on or impregnated onto a nasal and/or vaginal applicator and applied; being applied by a controlled-release mechanism; being applied as a liposome; or being applied on a polymer.
  • a subject e.g., mucosally (e.g., nasal mucosa, vaginal mucosa, etc.)
  • multiple methods including, but not limited to: being suspended in a solution and applied to a surface; being suspended
  • immunogenic compositions described herein are administered mucosally (e.g., using standard techniques; See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995 (e.g., for mucosal delivery techniques, including intranasal, pulmonary, vaginal and rectal techniques), as well as European Publication No. 517,565 and Illum et al., J. Controlled Rel., 1994, 29:133-141 (e.g., for techniques of intranasal administration), each of which is hereby incorporated by reference in its entirety).
  • mucosally e.g., using standard techniques; See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995 (e.g., for mucosal delivery techniques, including intranasal, pulmonary, vaginal and rectal techniques), as well as European Publication No. 517,565 and Illum e
  • immunogenic compositions described herein are administered dermally or transdermally, using standard techniques (See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995).
  • the present invention is not limited by the route of administration.
  • Mucosal vaccination such as intranasal vaccination, may induce mucosal immunity not only in the nasal mucosa, but also in distant mucosal sites such as the genital mucosa (See, e.g., Mestecky, Journal of Clinical Immunology, 7:265-276, 1987). In addition to inducing mucosal immune responses, mucosal vaccination also induces systemic immunity (See, e.g., Example 10).
  • non-parenteral administration e.g., mucosal administration of vaccines
  • provides an efficient and convenient way to boost systemic immunity e.g., induced by parenteral or mucosal vaccination (e.g., in cases where multiple boosts are used to sustain a vigorous systemic immunity)).
  • a composition comprising a NE and an immunogen of the present invention may be used to protect or treat a subject susceptible to, or suffering from, disease by means of administering a composition of the present invention via a mucosal route (e.g., an oral/alimentary or nasal route).
  • a mucosal route e.g., an oral/alimentary or nasal route.
  • Alternative mucosal routes include intravaginal and intra-rectal routes.
  • a nasal route of administration is used, termed “intranasal administration” or “intranasal vaccination” herein.
  • Methods of intranasal vaccination are well known in the art, including the administration of a droplet or spray form of the vaccine into the nasopharynx of a subject to be immunized.
  • a nebulized or aerosolized composition comprising a NE and immunogen.
  • Enteric formulations such as gastro resistant capsules for oral administration, suppositories for rectal or vaginal administration also form part of this invention.
  • Immunogenic compositions described herein may also be administered via the oral route.
  • a composition comprising a NE and an immunogen may comprise a pharmaceutically acceptable excipient and/or include alkaline buffers, or enteric capsules.
  • Formulations for nasal delivery may include those with dextran or cyclodextran and saponin as an adjuvant.
  • compositions described herein may also be administered via a vaginal route.
  • a composition comprising a NE and an immunogen may comprise pharmaceutically acceptable excipients and/or emulsifiers, polymers (e.g., CARBOPOL), and other known stabilizers of vaginal creams and suppositories.
  • compositions of the present invention are administered via a rectal route.
  • a composition comprising a NE and an immunogen may comprise excipients and/or waxes and polymers known in the art for forming rectal suppositories.
  • the same route of administration (e.g., mucosal administration) is chosen for both a priming and boosting vaccination.
  • multiple routes of administration are utilized (e.g., at the same time, or, alternatively, sequentially) in order to stimulate an immune response (e.g., using an immunogenic compositions described herein).
  • a composition comprising a NE and an immunogen is administered to a mucosal surface of a subject in either a priming or boosting vaccination regime.
  • a composition comprising a NE and an immunogen is administered systemically in either a priming or boosting vaccination regime.
  • a composition comprising a NE and an immunogen is administered to a subject in a priming vaccination regimen via mucosal administration and a boosting regimen via systemic administration.
  • a composition comprising a NE and an immunogen is administered to a subject in a priming vaccination regimen via systemic administration and a boosting regimen via mucosal administration.
  • systemic routes of administration include, but are not limited to, a parenteral, intramuscular, intradermal, transdermal, subcutaneous, intraperitoneal or intravenous administration.
  • a composition comprising a NE and an immunogen may be used for both prophylactic and therapeutic purposes.
  • immunogenic compositions described herein are administered by pulmonary delivery.
  • immunogenic compositions described herein can be delivered to the lungs of a subject (e.g., a human) via inhalation (e.g., thereby traversing across the lung epithelial lining to the blood stream (See, e.g., Adjei, et al. Pharmaceutical Research 1990; 7:565-569; Adjei, et al. Int. J. Pharmaceutics 1990; 63:135-144; Braquet, et al. J. Cardiovascular Pharmacology 1989 143-146; Hubbard, et al. (1989) Annals of Internal Medicine, Vol. III, pp.
  • an immunogenic composition described herein may be used to protect and/or treat a subject susceptible to, or suffering from, a disease by means of administering a compositions comprising a NE and an immunogen by mucosal, intramuscular, intraperitoneal, intradermal, transdermal, pulmonary, intravenous, subcutaneous or other route of administration described herein under conditions that induce an immunogen-specific immune response in the subject.
  • Methods of systemic administration of the vaccine preparations may include conventional syringes and needles, or devices designed for ballistic delivery of solid vaccines (See, e.g., WO 99/27961, hereby incorporated by reference), or needleless pressure liquid jet device (See, e.g., U.S. Pat. No.
  • the present invention may also be used to enhance the immunogenicity of antigens applied to the skin (transdermal or transcutaneous delivery, See, e.g., WO 98/20734; WO 98/28037, each of which are hereby incorporated by reference).
  • the present invention provides a delivery device for systemic administration, pre-filled with the vaccine composition of the present invention.
  • the present invention is not limited by the type of subject administered (e.g., in order to stimulate an immune response (e.g., in order to generate protective immunity (e.g., mucosal and/or systemic immunity))) a composition of the present invention. Indeed, a wide variety of subjects are contemplated to be benefited from administration of a composition of the present invention.
  • the subject is a human.
  • human subjects are of any age (e.g., adults, children, infants, etc.) that have been or are likely to become exposed to a microorganism (e.g., bacteria of the genus Burkholderia ).
  • the human subjects are subjects that are more likely to suffer from opportunistic infection (e.g., a subject with CF or an immune suppressed subject (e.g., a subject with human immunodeficiency virus)).
  • the subjects are non-human mammals (e.g., pigs, cattle, goats, horses, sheep, or other livestock; or mice, rats, rabbits or other animal).
  • compositions and methods of the present invention are utilized in research settings (e.g., with research animals).
  • the present invention provides a method to elicit an immune response (e.g., protective immune response) in infants (e.g., from about 0-2 years old) by administering to the infant a safe and effective amount of an immunogenic composition of the invention (e.g., a pediatric vaccine).
  • an immunogenic composition of the invention e.g., a pediatric vaccine.
  • Further embodiments of the invention include the provision of the immunogenic Burkholderia nanoemulsion compositions of the invention for use in medicine and the use of the Burkholderia nanoemulsion compositions of the invention in the manufacture of a medicament for the prevention (or treatment) of disease caused by Burkholderia .
  • the present invention is provides a method to elicit an immune response (e.g., a protective immune response) in the elderly population (e.g., in a subject 50 years or over in age, typically over 55 years and more generally over 60 years) by administering a safe and effective amount of an immunogenic composition of the invention.
  • an immune response e.g., a protective immune response
  • the elderly population e.g., in a subject 50 years or over in age, typically over 55 years and more generally over 60 years
  • Immunogenic compositions described herein may be formulated for administration by any route, such as mucosal, oral, topical, parenteral or other route described herein.
  • the compositions may be in any one or more different forms including, but not limited to, tablets, capsules, powders, granules, lozenges, foams, creams or liquid preparations.
  • Topical formulations may be presented as, for instance, ointments, creams or lotions, foams, and aerosols, and may contain appropriate conventional additives such as preservatives, solvents (e.g., to assist penetration), and emollients in ointments and creams.
  • Topical formulations may also include agents that enhance penetration of the active ingredients through the skin.
  • agents include a binary combination of N-(hydroxyethyl)pyrrolidone and a cell-envelope disordering compound, a sugar ester in combination with a sulfoxide or phosphine oxide, and sucrose monooleate, decyl methyl sulfoxide, and alcohol.
  • surfactants or wetting agents including, but not limited to, polyoxyethylene sorbitan mono-oleoate (Polysorbate 80); sorbitan mono-oleate (Span 80); p-isooctyl polyoxyethylene-phenol polymer (Triton WR-1330); polyoxyethylene sorbitan tri-oleate (Tween 85); dioctyl sodium sulfosuccinate; and sodium sarcosinate (Sarcosyl NL-97); and other pharmaceutically acceptable surfactants.
  • surfactants or wetting agents including, but not limited to, polyoxyethylene sorbitan mono-oleoate (Polysorbate 80); sorbitan mono-oleate (Span 80); p-isooctyl polyoxyethylene-phenol polymer (Triton WR-1330); polyoxyethylene sorbitan tri-oleate (Tween 85); dioctyl sodium sulfosuccinate; and sodium sarcosinate (Sar
  • compositions may further comprise one or more alcohols, zinc-containing compounds, emollients, humectants, thickening and/or gelling agents, neutralizing agents, and surfactants.
  • Water used in the formulations is preferably deionized water having a neutral pH.
  • Additional additives in the topical formulations include, but are not limited to, silicone fluids, dyes, fragrances, pH adjusters, and vitamins.
  • Topical formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation.
  • the ointment base can comprise one or more of petrolatum, mineral oil, ceresin, lanolin alcohol, panthenol, glycerin, bisabolol, cocoa butter and the like.
  • Immunogenic compositions described herein may additionally contain other adjunct components conventionally found in pharmaceutical compositions.
  • the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipuritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • additional materials useful in physically formulating various dosage forms of the compositions of the present invention such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • such materials when added, preferably do not unduly interfere with the biological activities of the immunogenic compositions described herein.
  • the formulations can be sterilized and, if desired, mixed with auxiliary agents (e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like) that do not deleteriously interact with the NE and immunogen of the formulation.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like
  • immunogenic compositions described herein are administered in the form of a pharmaceutically acceptable salt.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include, but are not limited to, acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v).
  • Suitable preservatives may include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • a composition comprising a NE and an immunogen is co-administered with one or more antibiotics.
  • one or more antibiotics may be administered with, before and/or after administration of a composition comprising a NE and an immunogen.
  • the present invention is not limited by the type of antibiotic co-administered.
  • these agents include agents that inhibit cell wall synthesis (e.g., penicillins, cephalosporins, cycloserine, vancomycin, bacitracin); and the imidazole antifungal agents (e.g., miconazole, ketoconazole and clotrimazole); agents that act directly to disrupt the cell membrane of the microorganism (e.g., detergents such as polmyxin and colistimethate and the antifungals nystatin and amphotericin B); agents that affect the ribosomal subunits to inhibit protein synthesis (e.g., chloramphenicol, the tetracyclines, erythromycin and clindamycin); agents that alter protein synthesis and lead to cell death (e.g., aminoglycosides); agents that affect nucleic acid metabolism (e.g., the rifamycins and the quinolones); the antimetabolites (e.g., trimethoprim and sulf
  • the present invention also includes methods involving co-administration of a composition comprising a NE and an immunogen with one or more additional active and/or immunostimulatory agents (e.g., a composition comprising a NE and a different immunogen, an antibiotic, anti-oxidant, etc.).
  • additional active and/or immunostimulatory agents e.g., a composition comprising a NE and a different immunogen, an antibiotic, anti-oxidant, etc.
  • additional active and/or immunostimulatory agents e.g., a composition comprising a NE and a different immunogen, an antibiotic, anti-oxidant, etc.
  • additional active and/or immunostimulatory agents e.g., a composition comprising a NE and a different immunogen, an antibiotic, anti-oxidant, etc.
  • the agents may be administered concurrently or sequentially.
  • the compositions described herein are administered prior to the other active agent(s).
  • the pharmaceutical formulations and modes of administration may be any of those described herein
  • the two or more co-administered agents may each be administered using different modes (e.g., routes) or different formulations.
  • the additional agents to be co-administered e.g., antibiotics, adjuvants, etc.
  • a composition comprising a NE and immunogen is administered to a subject via more than one route.
  • a subject that would benefit from having a protective immune response (e.g., immunity) towards a pathogenic microorganism may benefit from receiving mucosal administration (e.g., nasal administration or other mucosal routes described herein) and, additionally, receiving one or more other routes of administration (e.g., parenteral or pulmonary administration (e.g., via a nebulizer, inhaler, or other methods described herein).
  • administration via mucosal route is sufficient to induce both mucosal as well as systemic immunity towards an immunogen or organism from which the immunogen is derived.
  • administration via multiple routes serves to provide both mucosal and systemic immunity.
  • a subject administered a composition of the present invention via multiple routes of administration e.g., immunization (e.g., mucosal as well as airway or parenteral administration of a composition comprising a NE and immunogen of the present invention) may have a stronger immune response to an immunogen than a subject administered a composition via just one route.
  • Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides
  • hydrogel release systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides
  • sylastic systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides
  • peptide based systems such as fatty acids
  • wax coatings such as those described in U.S. Pat. Nos.
  • a composition comprising a NE and an immunogen of the present invention comprises a suitable amount of the immunogen to induce an immune response in a subject when administered to the subject.
  • the immune response is sufficient to provide the subject protection (e.g., immune protection) against a subsequent exposure to the immunogen or the microorganism (e.g., bacteria of the genus Burkholderia ) from which the immunogen was derived.
  • the present invention is not limited by the amount of immunogen used.
  • the amount of immunogen (e.g., Burkholderia bacteria (e.g., Burkholderia cepacia ) or one or more component parts thereof) in a composition comprising a NE and immunogen (e.g., for use as an immunization dose) is selected as that amount which induces an immunoprotective response without significant, adverse side effects.
  • the amount will vary depending upon which specific immunogen or combination thereof is/are employed, and can vary from subject to subject, depending on a number of factors including, but not limited to, the species, age and general condition (e.g., health) of the subject, and the mode of administration.
  • each dose (e.g., of a composition comprising a NE and an immunogen (e.g., administered to a subject to induce an immune response (e.g., a protective immune response (e.g., protective immunity))) comprises about 0.05-5000 ⁇ g of immunogen (e.g., recombinant and/or purified Burkholderia OMP protein).
  • immunogen e.g., recombinant and/or purified Burkholderia OMP protein
  • each dose (e.g., of a composition comprising a NE and an immunogen (e.g., administered to a subject to induce an immune response (e.g., a protective immune response (e.g., protective immunity))) comprises about 0.05-5000 ⁇ g of the immunogen (e.g., recombinant and/or purified Burkholderia OMP protein) and comprises about 0.05-5000 ⁇ g of another immunogen (e.g., recombinant and/or purified protein, adjuvant (e.g., cholera toxin), etc.).
  • the immunogen e.g., recombinant and/or purified Burkholderia OMP protein
  • another immunogen e.g., recombinant and/or purified protein, adjuvant (e.g., cholera toxin), etc.
  • each dose will comprise 1-500 ⁇ g, in some embodiments, each dose will comprise 350-750 ⁇ g, in some embodiments, each dose will comprise 50-200 ⁇ g, in some embodiments, each dose will comprise 25-75 ⁇ g of immunogen (e.g., recombinant and/or purified protein (e.g., Burkholderia antigen).
  • each dose comprises an amount of the immunogen sufficient to generate an immune response. An effective amount of the immunogen in a dose need not be quantified, as long as the amount of immunogen generates an immune response in a subject when administered to the subject.
  • each dose e.g., of a composition comprising a NE and an immunogen (e.g., administered to a subject to induce and immune response)
  • each dose is from 0.001 to 15% or more (e.g., 0.001-10%, 0.5-5%, 1-3%, 2%, 6%, 10%, 15% or more) by weight immunogen (e.g., neutralized bacteria, or recombinant and/or purified protein).
  • an initial or prime administration dose contains more immunogen than a subsequent boost dose
  • a composition comprising a NE and an immunogen of the present invention is formulated in a concentrated dose that can be diluted prior to administration to a subject.
  • dilutions of a concentrated composition may be administered to a subject such that the subject receives any one or more of the specific dosages provided herein.
  • dilution of a concentrated composition may be made such that a subject is administered (e.g., in a single dose) a composition comprising 0.5-50% of the NE and immunogen present in the concentrated composition.
  • a subject is administered in a single dose a composition comprising 1% of the NE and immunogen present in the concentrated composition.
  • Concentrated compositions are contemplated to be useful in a setting in which large numbers of subjects may be administered a composition of the present invention (e.g., an immunization clinic, hospital, school, etc.).
  • a composition comprising a NE and an immunogen of the present invention e.g., a concentrated composition
  • a composition comprising a NE and an immunogen of the present invention is stable at room temperature for more than 1 week, in some embodiments for more than 2 weeks, in some embodiments for more than 3 weeks, in some embodiments for more than 4 weeks, in some embodiments for more than 5 weeks, and in some embodiments for more than 6 weeks.
  • the emulsion compositions of the invention will comprise at least 0.001% to 100%, preferably 0.01 to 90%, of emulsion per ml of liquid composition. It is envisioned that the formulations may comprise about 0.001%, about 0.0025%, about 0.005%, about 0.0075%, about 0.01%, about 0.025%, about 0.05%, about 0.075%, about 0.1%, about 0.25%, about 0.5%, about 1.0%, about 2.5%, about 5%, about 7.5%, about 10%, about 12.5%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or about 98% of emulsion per ml of liquid composition. It should be understood that a range between any two figures listed above is specifically contemplated to be encompassed within the metes and bounds of the present invention. Some variation in dosage will necessarily occur depending on the condition of the specific pathogen and
  • a subject may receive one or more boost administrations (e.g., around 2 weeks, around 3 weeks, around 4 weeks, around 5 weeks, around 6 weeks, around 7 weeks, around 8 weeks, around 10 weeks, around 3 months, around 4 months, around 6 months, around 9 months, around 1 year, around 2 years, around 3 years, around 5 years, around 10 years) subsequent to a first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, and/or more than tenth administration.
  • boost administrations e.g., around 2 weeks, around 3 weeks, around 4 weeks, around 5 weeks, around 6 weeks, around 7 weeks, around 8 weeks, around 10 weeks, around 3 months, around 4 months, around 6 months, around 9 months, around 1 year, around 2 years, around 3 years, around 5 years, around 10 years
  • reintroduction of an immunogen in a boost dose enables vigorous systemic immunity in a subject.
  • the boost can be with the same formulation given for the primary immune response, or can be with a different formulation that contains the immunogen.
  • the dosage regimen will also, at least in part, be determined by the need of the subject and be dependent on the judgment of a practitioner.
  • Dosage units may be proportionately increased or decreased based on several factors including, but not limited to, the weight, age, and health status of the subject. In addition, dosage units may be increased or decreased for subsequent administrations (e.g., boost administrations).
  • compositions and methods of the present invention will find use in various settings, including research settings.
  • compositions and methods of the present invention also find use in studies of the immune system (e.g., characterization of adaptive immune responses (e.g., protective immune responses (e.g., mucosal or systemic immunity))).
  • Uses of the compositions and methods provided by the present invention encompass human and non-human subjects and samples from those subjects, and also encompass research applications using these subjects.
  • Compositions and methods of the present invention are also useful in studying and optimizing nanoemulsions, immunogens, and other components and for screening for new components. Thus, it is not intended that the present invention be limited to any particular subject and/or application setting.
  • compositions of the present invention are useful for preventing and/or treating a wide variety of diseases and infections caused by viruses, bacteria, parasites, and fungi, as well as for eliciting an immune response against a variety of antigens.
  • the compositions can also be used in order to prepare antibodies, both polyclonal and monoclonal (e.g., for diagnostic purposes), as well as for immunopurification of an antigen of interest.
  • polyclonal antibodies are desired, a selected mammal, (e.g., mouse, rabbit, goat, horse, etc.) can be immunized with the compositions of the present invention. The animal is usually boosted 2-6 weeks later with one or more—administrations of the antigen. Polyclonal antisera can then be obtained from the immunized animal and used according to known procedures (See, e.g., Jurgens et al., J. Chrom. 1985, 348:363-370).
  • the present invention provides a kit comprising a composition comprising a NE and an immunogen.
  • the kit further provides a device for administering the composition.
  • the present invention is not limited by the type of device included in the kit.
  • the device is configured for nasal application of the composition of the present invention (e.g., a nasal applicator (e.g., a syringe) or nasal inhaler or nasal mister).
  • a kit comprises a composition comprising a NE and an immunogen in a concentrated form (e.g., that can be diluted prior to administration to a subject).
  • kits are present within a single container (e.g., vial or tube).
  • each kit component is located in a single container (e.g., vial or tube).
  • one or more kit component are located in a single container (e.g., vial or tube) with other components of the same kit being located in a separate container (e.g., vial or tube).
  • a kit comprises a buffer.
  • the kit further comprises instructions for use.
  • a nanoemulsion composition of the present invention was tested to determine if pulmonary administration of the composition to a subject would elicit any histological changes and/or pathology.
  • a solution comprising 20% nanoemulsion (W 80 5EC) and 0.1% EDTA was administered via a nebulizer.
  • a custom murine nose only nebulization chamber with PARI LC nebulizer (Midlothian, Va.) and compressor was used.
  • a 0.9% NaCl solution was used as a control.
  • mice were administered nebulized nanoemulsion+EDTA or NaCl solution for 10 minutes. Twenty-four hours after administration, mice were sacrificed and physical properties and histology assessed.
  • composition comprising nanoemulsion (W 80 5EC) alone or in combination with EDTA and/or the presence of a hypertonic salt solution would be able to attenuate growth and/or kill bacteria found in the respiratory tract.
  • An overnight culture of Burkholderia cepacia or Pseudomonas aeruginosa was started in 6 ml of cation adjusted Mueller Hinton Broth (MHB) from a frozen bacterial stock at ⁇ 80° C.
  • the culture was incubated in a shaking incubator at 37° C.
  • bacteria were brought to logarithmic growth phase by back diluting the overnight culture 1:4 with fresh MHB.
  • Back diluted culture was incubated at 37° C. in the shaking incubator until the OD600 reached between 0.40 to 0.45.
  • One ml of the culture was spun down at 3500 to 4000 rpm for 15 minutes.
  • the reaction was diluted with 500 ⁇ l of 1 ⁇ PBS and vortexed to mix the contents. The contents were centrifuged at 4000 rpm for 15 minutes to pellet the bacteria and separate the nanoemulsion. Supernatant containing the nanoemulsion was removed and the bacterial pellet resuspended in 1 ⁇ PBS. Undiluted or dilutions of the resuspended bacterial pellet was plated onto LB agar plates for colony count. Log killing of the bacteria were calculated from ratio with the starting numbers.
  • Nanoemulsion alone or in combination with EDTA was able to achieve complete killing of 10 6 bacteria in PBS in 60 minutes.
  • EDTA e.g., 10-20 mM EDTA
  • hypertonic saline e.g., 6-7% NaCl
  • the killing ability of the nanoemulsion was strikingly enhanced, achieving complete killing within 15 minutes while in the presence of 20 mM EDTA.
  • nanoemulsions comprising a lower concentration of EDTA were able to achieve complete killing of bacteria in 30 minutes in the presence of hypertonic saline.
  • the present invention provides that a nanoemulsion composition comprising EDTA can be used to kill (e.g., completely) bacteria over a short time period (e.g., ⁇ 60 minutes). Moreover, the present invention provides that nanoemulsion with hypertonic saline and EDTA can be used to kill bacteria over even shorter time periods (e.g., ⁇ 30 minutes, ⁇ 15 minutes) and that the concentration of EDTA and hypertonic saline solution alter the pace at which the nanoemulsion is able to eradicate the bacteria. The present invention also demonstrates that compositions of the present invention are able to eradicate a mixed population of bacteria.
  • pyrrocinia HI3642 BcRLR collection
  • the clinical type strains B. cenocepacia LMG 16656 T aka J2315
  • Nanoemulsion P 407 5EC was manufactured by NANOBIO Corp. (Ann Arbor, Mich.). P 407 5EC droplets had a mean particle diameter of 400 nm. Surfactants and food substances utilized in the manufacture of P 407 5EC were ‘Generally Recognized as Safe’ (GRAS) by the FDA, and manufactured in accordance with Good Manufacturing Practices (GMP). The concentration of CPC was used as a surrogate for the amount of P 407 5EC used experimentally. P 407 5EC was stable for no less than 12 months at 40° C.
  • GRAS Generally Recognized as Safe
  • GMP Good Manufacturing Practices
  • P 407 5EC is opaque, the MICs of this compound for test bacteria were determined by using a modification of the Clinical and Laboratory Standards Institute (CLSI)-approved microtiter serial dilution method (See, e.g., Clinical and Laboratory Standards Institute. 2006. Approved standard M7-A7, seventh edition. Clinical and Laboratory Standards Institute, Wayne, Pa.).
  • CPC Clinical and Laboratory Standards Institute
  • P 407 5EC was diluted to a concentration of 2 mg/ml (of CPC) in MH broth supplemented with 7% NaCl and 20 mM EDTA. Serial two-fold dilutions of this preparation were made in unsupplemented MH broth and aliquoted into 96-well flat bottom microtiter plates (100 ⁇ l/well).
  • Bacteria from overnight growth on MH agar were suspended in MH broth to a 0.5 McFarland turbidity standard (absorbance of 0.08-0.13 at 625 nm), further diluted 1:100 in MH broth, and added (5 ⁇ l/well) to the P 407 5EC serial dilution wells. Appropriate controls, including wells with bacteria but no P 407 5EC and wells with P 407 5EC dilutions but no bacteria, were included on each plate.
  • Microtiter plates were shaken briefly, and 1 ⁇ l was removed from wells containing bacteria but no P 407 5EC, diluted in 1 ml of MH broth, plated on MH agar (100 ⁇ l), and incubated for 24-48 h at 37° C. to determine bacterial concentrations of initial inoculums. Microtiter plates were then incubated at 37° C. without shaking. To determine MBCs, 10 ⁇ l were removed from each well after overnight growth, spotted on MH agar, and incubated at 37° C. Colonies were enumerated 24 h later, and MBCs were recorded as the P 407 5EC concentration with a 3 log decrease in CFU/ml compared to the initial inoculum.
  • MICs 10 ⁇ l of resazurin (R&D SYSTEMS, Minneapolis, Minn.) were added to each well and microtiter plates were shaken briefly, covered with foil, and incubated at 37° C. without shaking. Wells were visually inspected the next day, and the MIC was recorded as the lowest concentration of P 407 5EC remaining a blue color. MIC results were further quantified by recording fluorescence on a spectrofluorometer at 560 nm excitation/590 nm emission.
  • Biofilm growth and susceptibility testing To identify biofilm-forming isolates, bacteria were grown overnight in tryptic soy broth, adjusted to a 0.5 McFarland turbidity standard, and further diluted 1:10 in MH broth, and 100 ⁇ l were seeded in triplicate in internal wells of 96-well flat bottom microtiter plates. Negative control wells without bacteria were included. To minimize evaporation, the remaining wells were filled with MH broth, and the plate was wrapped with plastic wrap before incubating for 48 h at 37° C. without shaking. Wells were gently washed twice with phosphate buffered saline (PBS; pH 7.4), dried for 2 h at 37° C., and then stained with 1% crystal violet in water for 15 min.
  • PBS phosphate buffered saline
  • biofilm was resuspended in the same wells used for MBIC testing by shaking the microtiter plate and then scraping the sides of the wells with a pipet tip. Ten ⁇ L were removed from each well, spotted on MH agar plates and incubated at 37° C. After overnight growth, MBECs were recorded as the lowest P 407 5EC concentration resulting in no growth (See, e.g., Tomlin et al., Can J Microbiol. 2001 October; 47(10):949-54).
  • the MIC results are shown in FIG. 11 . All strains were inhibited by the concentrations of P 407 5EC tested. The MIC 50 for the entire panel of 150 strains was 31.2 ⁇ g/ml; the MIC 90 was 125 ⁇ g/ml. Thirty eight strains (25%) were inhibited by the lowest concentration of P 407 5EC tested (15.6 ⁇ g/ml), and only a single strain each required a concentration of 250 ⁇ g/ml and 500 ⁇ g/ml for inhibition.
  • P 407 5EC was slightly more active against non- Burkholderia strains (MIC 50 , 31.2 ⁇ g/ml; MIC 90 62.5 ⁇ g/ml) than Burkholderia strains (MIC R , 62.5 ⁇ g/ml; MIC 90 125 ⁇ g/ml) (See FIG. 9 ). Activity was comparable across the 10 Burkholderia species tested. No difference was found in the activity of P 407 5EC against multi-drug resistant Burkholderia strains compared to strains susceptible to one or more antibiotics.
  • MBCs were determined on a subset of 34 strains including 22 Burkholderia and 12 non- Burkholderia . All MBCs were within 1 dilution of the respective MICs for this subset of strains except for a single B. cenocepacia strain that had a MIC of 250 ⁇ g/ml and an MBC of 2000 ⁇ g/ml. A time-kill study of B.
  • multivorans ATCC 17616 showed time- and concentration-dependent killing, with a 99% decrease in bacterial viability within 90 min at a P 407 5EC concentration two times greater than the MIC and complete killing within 30 min at concentrations eight times greater than the MIC (See FIG. 10A ).
  • a composition comprising nanoemulsion P 407 5EC, 7% saline and 20 mM EDTA was tested for its ability to kill bacteria present in biofilms (individual or mixed).
  • Biofilm Growth Bacterial biofilms on polycarbonate membranes were formed following the procedure described in Current Protocols in Microbiology (John Wiley & Sons, Inc., NJ, USA). Polycarbonate membranes were sterilized by exposing the membrane to UV light for 10 minutes each side using sterile forceps or by steam sterilization at 121° C. for 20 minutes. The sterile membrane was placed on the surface of an agar medium with the shiny surface facing upwards. Overnight culture of the bacterial strain of interest was diluted with broth media to an OD 600 of 0.05 and 0.5 ⁇ l of the diluted culture placed onto the shiny surface of the polycarbonate membrane on the agar plate medium. The plate with the membrane was incubated at 37° C. for 24 hours following which; the membrane is transferred to a new agar medium for another 24 hours for biofilm formation.
  • This example describes the effectiveness of various nanoemulsion formulations against various gram negative pathogens that may be associated with CF, wounds, burns and burn patients, or other environments.
  • CF is characterized by chronic respiratory infection that begins early in life with Staphylococcus aureus and Haemophilus influenzae infections and later colonization with mucoid strains of Pseudomonas aeruginosa .
  • early colonization with Burkholderia cepacia correlates with a poor prognosis in CF patients.
  • Patients are often prescribed inhaled tobramycin to prevent exacerbations of bacteria infections. With time, patients can become unresponsive to tobramycin therapy/prophylaxis. New inhaled topical agents that are not cross-resistant to known antibiotics would be valued as an alternative to inhaled tobramycin.
  • nanoemulsions were evaluated for microbiological activity against gram-negative isolates, including P. aeruginosa and B. cepacia . All 3 nanoemulsions kill quickly in the presence of an outer membrane permeabilizer such as EDTA. The nanoemulsions were evaluated against 23 of the 35 gram-negative isolates for cidal activity, and all were bactericidal (85-95%). Thus, the one or more of these novel nanoemulsions can be used to prevent exacerbation of chronic pulmonary infections.
  • MICs and MBCs were determined using CLSI standard methods. MICs to nanoemulsions were determined in the presence of 5 mM EDTA, a known enhancer of nanoemulsion activity. The addition of alamar blue, a redeox indicator that yields a colorimetric change in response to metabolic activity, was used to determine the MICs of nanoemulsions that are opaque at higher concentrations.
  • Nanoemulsions W 20 5EC, P 407 5EC, and W 20 5GBA 2 ED are oil-in-water emulsions manufactured from ingredients that are Generally Recognized As Safe (GRAS) with a cationic detergent (cetylpyridinium chloride (CPC) or benzalkonium chloride (BA)) as active ingredients that have proven safe for oral human use.
  • GRAS Generally Recognized As Safe
  • CPC cetylpyridinium chloride
  • BA benzalkonium chloride
  • the emulsion is formed from highly purified oil, ethanol, a nonionic surfactant and water.
  • the average nanoemulsion droplet size was 180 nm for W 20 5EC and 350 nm for P 407 5EC and W 20 5GBA 2 ED, as measured by dynamic light scattering using a Malvern Zetasizer Nano3600 (Malvern Instruments Ltd., Worcestershire, UK).
  • the formulations for W 20 5EC and P 407 5EC are described above.
  • W 20 5GBA 2 ED (v/v %) is distilled water (18.93%), TWEEN 20 (5%), Glycerine (8%), Soybean oil (64%), BTC 824 50% (4%) (Stepan, Northfield, Ill.), and EDTA (0.0745%).
  • the formulation may be diluted in water (e.g., 60% W 20 5GBA 2 ED, 40% water) to provide 60% W 20 5GBA 2 ED.
  • This material may be diluted to a 10% formulation with 10 mM EDTA (e.g., 76 g water per 24 g 100 mM EDTA per 20 g 60% W 20 5GBA 2 ED).
  • 10 mM EDTA e.g., 76 g water per 24 g 100 mM EDTA per 20 g 60% W 20 5GBA 2 ED.
  • a formulation comprising oil, water, glycerine, surfactant, and BTC (n-alkyl dimethyl benzyl ammonium chloride), and/or EDTA is provided.
  • the source of the clinical isolates were blood stream or skin and soft tissue isolates collected by JMI Laboratories in the past 2 years.
  • MIC and MBC determinations were evaluated in cation-adjusted Mueller-Hinton broth by microdilution per M7-A7 (2006).
  • MICs for a panel of gram-negative isolates were determined for the nanoemulsions, W205EC, P4075EC, and W205GBA2ED in the presence of 5 mM EDTA.
  • EDTA was used to permeabilize the gram-negative envelope, aiding infusion of the nanodroplets to the cell membrane, resulting in lysis. Alamar blue was added to the assay panels 2 hours post-inoculation for enhanced MIC endpoint detection.
  • MBC values were assessed for all nanoemulsions and a comparator compound (fusidicacid) by plating the entire broth content from the MIC well and from those four doubling dilutions above the MIC for each selected organism onto blood agar growth media. Quantitative colony counts were performed on the initial inoculum. The lowest concentration of antimicrobial agent that killed ⁇ 9.9% of the starting test inoculum was defined as the MBC endpoint. See Tables 2, 3, and 4.
  • Nanoemulsions were broadly active against Gram-negative species, including multidrug-resistant isolates. The documented MICs were within the range of concentrations achievable with topical application to skin or mucosal tissues. One or more of the nanoemulsions may be useful for prophylaxis and/or therapeutic treatment of chronic pulmonary infections in cystic fibrosis patients.
  • H. influenzae is an important respiratory pathogen implicated in acute exacerbations of cystic fibrosis patients as well as upper and lower respiratory tract infections for non-CF normal individuals.
  • This example determined the minimum inhibitory concentration of three exemplary nanoemulsions (W 20 5EC, P 407 5EC and W 20 5GBA 2 ) plus comparator drugs against clinical isolates of H. influenzae .
  • the results, as described in more detail below, show that P 407 5EC and W 20 5EC had similar MIC ranges of 1.88-3.75 ⁇ g/ml.
  • W 20 5GBA 2 had a MIC range of 8-16 ⁇ g/ml.
  • the addition of EDTA at 1 mM to each well containing nanoemulsion had no effect on the MIC. Concentrations of EDTA greater than 1 mM inhibited the growth of bacteria, likely reducing the concentration of cations to levels below those that support growth.
  • MBC data for the three nanoemulsions+1 mM EDTA were obtained for seven strains. The MBCs were within 4-fold of the respective MICs, consistent with each nanoemulsion having bactericidal activity against H. influenzae isolates.
  • W 20 5EC, lot x1151, at a concentration of 5000 ⁇ g/ml (stock concentration) was a pooled sample of clinical trial material (NB-001), manufactured at Patheon, Mississauga, Ontario, Canada.
  • P 407 5EC, lot X1138, and W 20 5GBA 2 , lot X1103, were manufactured at NanoBio Corporation.
  • Comparator compounds, azithromycin, ampicillin, cefepime and clindamycin were purchased from The United States Pharmacopeia; catalog numbers were 1046056, 1033000, 1097636, 1136002, respectively.
  • Tetracycline was purchased from Fluka Biochemika, catalog number 87128.
  • the emulsions were produced by mixing a water immiscible oil phase with an aqueous phase.
  • the base formulations are shown in Table 5 below and represent the neat emulsions, which were further diluted to the desired %. Compositions are w/w % unless otherwise noted.
  • H. influenzae Ten clinical isolates of H. influenzae were obtained from Case Western Reserve University, Cleveland, Ohio.
  • the ranges of drug concentrations tested were as follows: Azithromycin at 0.25-128 ⁇ g/ml, Ampicillin at 0.063-32 ⁇ g/ml, Cefepime at 0.063-32 ⁇ g/ml, Clindamycin at 0.125-64 ⁇ g/ml, Tetracycline at 0.125-64 ⁇ g/ml, P 407 5EC at 0.12-60 ⁇ g/ml cetylpyridinium chloride, W 20 5EC at 0.12-60 ⁇ g/ml cetylpyridinium chloride, W 20 5GBA 2 at 0.125-64 ⁇ g/ml benzalkonium chloride.
  • the MICs/MBCs are expressed as the concentration of the cationic surfactant; W 20 5EC and P 407 5EC both contain cetylpyridinium chloride (CPC) as the cationic surfactant, while W 20 5GBA 2 contains benzalkonium chloride (BA) as the cationic surfactant.
  • Isolates were also tested with the same range of nanoemulsion concentrations+1-5 mM EDTA to see if the latter enhanced the activity of the nanoemulsion. EDTA concentrations were tested alone to ensure that chelation of cations by EDTA was sufficient to inhibit bacterial growth independent of the nanoemulsion.
  • MIC Minimum inhibitory concentrations
  • MBC values were assessed for the nanoemulsions+1 mM EDTA for 7 clinical strains and the ATCC 49257 isolate by plating 10 ⁇ l from the well determined to be the MIC and 4 wells above the MIC onto chocolate blood agar media.
  • the lowest concentration of antimicrobial agent that killed ⁇ 99.9% of the starting test inoculum was defined as the MBC.
  • a compound or nanoemulsion is defined as bactericidal if its MBC/MIC ratio was ⁇ 4.
  • Results Data for the MICs and MBCs are shown in Tables 6-7.
  • P 407 5EC, W 20 5EC, and W 20 5GBA 2 were equally effective antimicrobials in the presence or absence of 1 mM EDTA.
  • the MIC 90 values for these compounds were 3.75, 3.75, and 16 ⁇ g/ml, respectively (Table 6).
  • MIC 90 values the majority of isolates were susceptible to azithromycin, cefepime, tetracycline and ampicillin, but were resistant to clindamycin and ampicillin.
  • the MBC values were within 4-fold of the respective MICs for each isolate tested (Table 7); thus each of the nanoemulsions were bactericidal for H. influenzae , with a range of MBCs for P 407 5EC, W 20 5EC, and W 20 5GBA 2 of 1.88-7.5, 1.88-7.5 and 8-16 ⁇ g/ml, respectively.
  • the MBC was relied upon to provide the MIC.
  • Table 8 contain plate counts for MBC.
  • the second number is the plate count of a 10 ⁇ l sample from the microtiter plate used to determine the MBC.
  • the MBC was defined as ⁇ 3-log reduction of the inoculum. The darker highlighting corresponds to MIC.
  • influenzae MIC of Nanoemulsions with EDTA Highlighted in Green are rows containing plate counts for the MBC. The second number is the plate count of a 10 ⁇ l sample from the microtiter plate used to determine the MBC. The MBC was defined as ⁇ 3-log reduction of the inoculum.
  • MRSA nanoemulsions
  • the purpose of this example was to evaluate 4 nanoemulsions according to the invention for use in the topical treatment of infectious diseases.
  • the nanoemulsions tested were W 80 5EC, W 20 5ECEDL2, W 20 5GBA 2 ED and P 407 5EC.
  • This example determined the minimum inhibitory concentration (MIC) of the four nanoemulsions plus comparator drugs against 29 clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA).
  • MRSA methicillin-resistant Staphylococcus aureus
  • the four nanoemulsions were (a) W 80 5EC, (b) W 20 5ECEDL2, (c) W 20 5GBA 2 ED and (d) P 407 5EC. See Table 10, which provides drug panel templates (with increasing dilutions across the panel).
  • the emulsions were produced by mixing a water immiscible oil phase with an aqueous phase.
  • the W 20 5ECEDL2 formulation was microfluidized.
  • the base formulations are shown in Table 9 below and represent the neat emulsions, which were further diluted to the desired %. Compositions are w/w % unless otherwise noted.
  • the ranges of drug concentrations were as follows: clindamycin (64-0.125 ⁇ g/ml), doxycycline (128-0.25 ⁇ g/ml), erythromycin (128-0.25 ⁇ g/ml), levofloxacin (64-0.125 ⁇ g/ml), mafenide acetate (128-0.25 ⁇ g/ml), mupirocin (64-0.125 ⁇ g/ml), silvadene (128-0.25 ⁇ g/ml), vancomycin (64-0.125 ⁇ g/m), oxacillin (16-0.03125 ⁇ g/ml), sulfamethoxazole (32-0.0625 ⁇ g/ml), trimethoprim (64-125 ⁇ g/ml), silver nitrate (0.01-0.00002%), W 80 5EC (32-0.0625 ⁇ g/ml), W 20 5ECEDL2 (32-0.0625 ⁇ g/ml), W 20 5GBA 2
  • Minimum inhibitory concentrations were determined visually as the first totally clear well in the series of the 10 concentrations of each drug. For the nanoemulsions, because they are intrinsically opaque, the MIC was called as the first well observed to have some clearing from the least concentrated to the most concentrated.
  • Minimum bactericidal concentrations (MBC) values were assessed for all the antimicrobials tested. The lowest concentration of antimicrobial agent that killed 2: 99.9% of the starting test inoculum was defined as the MBC.
  • a compound or nanoemulsion is defined as bactericidal if its MBC/MIC ratio was ⁇ 4.
  • Mupirocin is a topical treatment for skin and soft tissue infections and is also used to eradicate carriage of MRSA; 31% of the isolates were resistant to this antibiotic.
  • the MBC 90 values for all of the nanoemulsions were within four-fold of the respective MIC 90 values, indicating that the nanoemulsions are bactericidal to ⁇ 90% of the isolates.
  • vancomycin was bactericidal.
  • MIC analysis MIC ( ⁇ g/ml) Rank Maf. Order Clind Dox Eryth Levo Acet Mup Silva Vanco Ox Sulfa 29 >64 4 >128 >64 >128 >64 64 2 >16 >32 28 >64 4 >128 >64 >128 >64 64 1 >16 >32 27 >64 4 >128 32 >128 >64 32 1 >16 4 26 >64 4 >128 32 >128 >64 32 1 >16 4 25 >64 0.25 >128 32 >128 >64 32 1 >16 2 24 >64 0.25 >128 32 >128 >64 32 1 >16 2 23 >64 0.25 >128 32 >128 >64 32 1 >16 2 22 >64 0.25 >128 16 >128 >64 32 1 >16 1 21 >64 0.25 >128 16 >128 16 32 1 >16 1 20 >64 0.25 >128 16 >128 0.25 32 1 >16 1 19 >64 0.25 >
  • MBC analysis MBC ( ⁇ g/ml) Maf. Rank Order Clind Dox Eryth Levo Acet. Mup Silva. Vanco Ox Sulfa 29 >64 >128 >128 >64 >128 >64 >128 4 >16 >32 28 >64 >64 >128 >64 >128 4 >16 >32 27 >64 >64 >128 >64 >128 >64 >128 4 >16 >32 26 >64 >64 >128 >64 >128 2 >16 >32 25 >64 >64 >128 >64 >128 >64 >128 2 >16 >32 24 >64 >4 >128 >64 >128 >64 >128 2 >16 >32 23 >64 >4 >128 64 >128 >64 >128 2 >16 >32 22 >64 >4 >128 64 >128 >64 >128 2 >16 >16 21 >64 >4 >128 >32
  • W 80 5EC, W 20 5ECEDL2, W 20 5GBA 2 ED and P 407 5EC appeared equally effective against clinical isolates of MRSA, with MIC 90 values of 2 or 4 ⁇ g/ml.
  • the four nanoemulsions were bactericidal.
  • the MRSA were multidrug-resistant, emphasizing the need for new agents for treating MRSA infections. Since nanoemulsions according to the invention also have activity against serious gram-negative pathogens, such as Pseudomonas aeruginosa and Burkholderia spp.
  • the nanoemulsions are useful for inhalation treatment/maintenance of cystic fibrosis patients.
  • the nanoemulsions are useful in treating burn wounds to prevent/treat infections of the pathogenic agents described herein.
  • the purpose of this example was to determine the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) of an exemplary nanoemulsion (P 407 5EC) side by side with comparator drugs against clinical isolates of Pseudomonas aeruginosa, Burkholderia cenocepacia, Acinetobater baumanni, Stenotrophomonas maltophilia from patients suffering from cystic fibrosis (CF).
  • MIC minimum inhibitory concentration
  • MMC minimum bactericidal concentration
  • P 407 5EC+EDTA had an MIC range of ⁇ 4-16, ⁇ 4-64, 2-8, ⁇ 1-16 ⁇ g CPC/ml, respectively.
  • the addition of EDTA at 5 mM for the Pseudomonas and Burkholderia and 1 mM to Acinetobacter and Stenotrophomonas to each well containing nanoemulsion improved the MIC.
  • MBC data for the nanoemulsion+EDTA were obtained for 20 Pseudomonas, 10 Burkholderia, 10 Acinetobacter and 11 Stenotrophomonas . For three genera of bacteria tested, the data suggested bacteriostatic activity. The MBCs were within 4-fold of the respective MICs for the Stenotrophomonas , consistent with the nanoemulsion having bactericidal activity.
  • P 407 5EC+EDTA in combination with colistin was found to be indifferent, only 20% synergy in by the FIC and 0% by the FBC, as well as when in combination with tobramycin, only 10% synergy in by the FIC and 10% by the FBC.
  • P 407 5EC+EDTA in combination with colistin was found to be indifferent, only 30% synergy in by the FIC and 10% by the FBC, but when in combination with tobramycin, 50% synergy in by the FIC and 20% by the FBC.
  • Multidrug-resistant Gram-negative bacteria isolated from CF patients were tested in this example.
  • MICs and MBCs were determined using CLSI guidelines and standard methods M7-A7 and M100-S17. Depending on the genus, MICs were performed in the presence of 1 or 5 mM EDTA, a permeability enhancer of NE activity. Stenotrophomonas and Acintetobacter had 1 mMEDTA in the nanoemulsion. Pseudomonas and Burkholderia had 5 mM EDTA in the nanoemulsion. This was the case during the standard MIC and MBC determination as well as the Checkerboard synergy work and time kill study to be described below. The addition of alamar blue, a redox indicator that yields a colorimetric change in response to metabolic activity, was used to determine the MICs of the NE (P 407 5EC) because of opacity at higher concentrations.
  • the Fractional Inhibitory Concentration (FIC) Index examines the ratio of the MIC of a single drug when in combination with another to the MIC of that drug alone. The reduction of the MIC when a drug is in combination results in a fraction that is less than one.
  • Row D denotes the previously determined MIC used to chose the flanking concentrations of the two drugs being combined.
  • the horizontal slash marks on the X and Y axis indicate typical MIC concentration of each drug alone.
  • the time-kill study was done over a brief time curve of 10, 20, 30 minutes including a 0 minute non-treated control was conducted to fix samples for pictures under the electron microscope.
  • the composition of the P 407 -5EC is provided in Table 16, below.
  • the emulsion was produced by mixing a water immiscible oil phase with an aqueous phase.
  • the base formulation is below and represents the neat emulsion, which was further diluted to the desired %.
  • Table 17 provides details regarding the preparation of the 96-well drug panel plates
  • Table 18 provides details regarding the preparation of the 96-well checkerboard synergy plates.
  • Buffer 5.00 3200 17.22 Lot H0E040 pH 7.2 Levofloxacin- 361.37 999.00 16 1 ⁇ 2 volume 10.00 1600 16.02 Sigma 1333515 water, then 0.1 mol/L NaOH dropwise to dissolve Piperacillin- 535.57 984 1024 1 ⁇ 2 volume 2.00 102400 208.13 USP Lot H water, then 0.1 mol/L NaOH dropwise to dissolve Tobramycin- 467.52 970 32 water 5.00 3200 16.49 USP Lot L0E077 Cefoxitin-USP 449.44 992 256 1 ⁇ 2 volume 2.00 25600 51.61 Lot J0E038 water, then 0.1 mol/L NaOH dropwise to dissolve Ceftazidime-Lot H 535.57 852 32 Calcium 2.00 3200 7.51 Carbonate Sln 10% of the weight of compound to be dissolved YELLOW HIGHLIGHT INDICATES POTENCY TAKEN FROM PERCENT PURITY (99% 999 ug/mg).
  • the calculations provided a 2x stock that is diluted in broth from the start and through the nine 1/2 dilutions. A 1/50 dilution at any time is not required because the dilutions are made directly in broth.
  • a 1/50 dilution of the EDTA 100x stock was made in each well of the 12 well trough containing nanoemulsion. Only 10 wells of the 12 well trough contain drug, the last two wells are for broth only which set up the negative and positive growth controls.
  • the inoculated wells were observed after 20 hours of incubation at 35 degrees C.
  • each row there is a range from high to low concentrations of the drug respective of the row from left to right, each well 1 ⁇ 2 the concentration of the adjacent well to the left.
  • the first clear well in the series was called the MIC. This is the concentration of drug that inhibited the growth of the bacteria from turning the broth turbid.
  • alamar blue (CellTiter Blue, Promega) was added for the row containing nanoemulsion at a rate of 20 uL to 100 uL of culture volume.
  • the agar plates were incubated for 24 hours at 35 degrees C. and colonies were counted. The resulting colony counts were compared to the starting concentration of the inoculum to determine which well, that is which concentration of drug, resulted in a three log drop in cfu/mL.
  • the starting inoculum in each well was generally between 2-7 ⁇ 10 ⁇ 5 cfu/mL.
  • the specific concentration for starting inoculum was determined for each test plate by sampling the inoculum used to inoculate the microtiter plate at the time it was set up. Those exact concentrations of bacteria were used to determine the MBC for each drug in the panel for each test plate.
  • the MBC is defined as the concentration of drug that reduced the number of bacteria 99.9%, a three log drop from the original concentration of bacteria.
  • the MIC is the landmark used to begin sampling 10 uL/well from the MIC plus all wells through to column 11 to the right and put on agar plates, one sample per plate. The agar plates were incubated for 24 hours at 35 degrees C. and colonies were counted. The resulting colony counts were compared to the starting concentration of the inoculum to determine which well, that is which concentration of drug, resulted in a three log drop in cfu/mL.
  • the inoculated wells were observed after 20 hours of incubation at 35 degrees C.
  • each row there is a range from high to low concentrations of the drug respective of the row from right to left, each well 1 ⁇ 2 the concentration of the adjacent well to the left.
  • the rows contained the nanoemulsion, drug A. Going up and down in the columns is drug B and here is the second drug tested in combination going high to low from A to G, respectively.
  • Row H did not contain the second drug for it to act as the control for drug A.
  • Column 1 did not contain any drug A so that column 1 could be the control for drug B.
  • first clear well in the series from right to left in the rows was called the MIC.
  • the well with the MIC and the well with the MBC were noted for each row. All wells noted for the MIC of the two drugs in combination were compared to the MIC of concentration of drug A and drug B each acting alone. For example, a well that contained 2 ug/mL of drug A and 8 ug/mL of drug B would have the concentration of A at 1 ug/mL when in combination divided by 16 ug/mL when acting alone. This renders a ratio of 0.125. Further in this example, 8 ug/mL of drug B would be divided by 32 ug/mL of B acting alone. The ratio for drug B in combination:alone is then 0.25.
  • the FIC index is said to be 0.125 for drug A and 0.25 for drug B, the sum is 0.325 in this example.
  • the definition of synergy is for the sum to be equal to 0.5 or less. Indifference is greater than 0.5 to less than or equal to four. Antagonism is greater than four. See e.g., FIG. 19 for a detailed illustration.
  • a bacterial suspension of Burkholderia NB 29@O.D. 0.14-0.20 was made up.
  • 500 uL saline and 500 uL of the bacterial suspension was mixed for the TO, non-treated control, then mixed one to one with TA (Trypton-Azolectin) broth with 5 mM CaCl 2 to be diluted and plated as described below.
  • TA Tetra-Azolectin
  • a treatment tube added one to one the bacterial suspension to the treatment mix (2 ⁇ P4075EC at 64 ug/mL CPC and 10 mM EDTA) and after 10, 20 and 30 minutes drew 400 uL at each time point and mixed with 400 uL TA broth with 5 mM CaCl 2 .
  • 100 uL were drawn to be diluted serially 1/10 in saline to 10 ⁇ 8 .
  • 100 uL were then drawn from each dilution tube in the series and plated in triplicate. These plates were incubated at 35° C. overnight and counted to determine the cfu/mL at each time point.
  • a sample of 450 uL was added to 113 uL of glutaraldehyde for each respective time point.
  • Samples were allowed to evaporate in the hood overnight 8 Samples were mounted on SEM stubs, using the mixture of Colloidal graphite and duco cement 9 Samples were placed in a vacuum desiccator overnight 10 Samples were sputter-coated with gold using “Polaron” sputter coater 11 Samples were examined on an “Amray 1910 FE” Sacnning Electron Microscope and digitally imaged using “Xstream” imaging software
  • the MIC 90 /MBC 90 values for P 407 5EC were 8/64 ⁇ g/ml for P. aeruginosa, 64/>514 ⁇ g/ml for B. cenocepacia, 8/64 ⁇ g/ml for A. baumannii and 8/32 ⁇ g/ml for S. maltophilia .
  • Colistin had MIC 90 /MBC 90 values of 2/8, >32/>32, 11>16 and >32/>32 for P. aeruginosa, B. cenocepacia, A. baumannii and S. maltophilia , respectively.
  • Cefepime, imipenem, levofloxacin and tobramycin had MIC 90 /MBC 90 values of ⁇ 32/>32, ⁇ 32/>32, 16/16 and >32/>32 ⁇ g/ml, respectively, against all strains.
  • P 407 5EC+EDTA in combination with colistin was found to be indifferent, only 20% synergy in by the FIC and 0% by the FBC, as well as when in combination with tobramycin, only 10% synergy in by the FIC and 10% by the FBC.
  • P 407 5EC+EDTA in combination with colistin was found to be indifferent, only 30% synergy in by the FIC and 10% by the FBC, but when in combination with tobramycin, 50% synergy in by the FIC and 20% by the FBC.
  • Time-kill resulted in an overall 4.44 log reduction in cfu/ml from the untreated beginning to the 30 minute time point.
  • Each 10 minute time point had between 1-2 log reduction as follows: from the untreated to the 10 minute point there was a 1.40 log reduction, from the 10 to 20 minute time points there was a 1.91 log reduction and from the to 30 minute time points there was a 1.13 log reduction. See FIGS. 20A-20D .
  • nanoemulsions such as the tested P 407 5EC
  • the SEM images demonstrate the kill on contact mechanism, here in this case a Gram negative bacterium with a reputation of having a tough outer membrane.
  • Topical Nanoemulsion Therapy Reduces Bacterial Wound Infection and Inflammation Following Burn Injury
  • Rats Male specific pathogen-free Sprague-Dawley rats (Harlan, Indianapolis, Ind.) weighing approximately 250-300 g were used in all experiments. Rats were housed in standard cages and allowed to acclimate to their surroundings for 7 days prior to being used in experiments. They were kept on a 12 hour light cycle and provided with unrestricted access to standard rat chow and water throughout the study. Experiments were performed in accordance with National Institutes of Health guidelines for care and use of animals. Approval for the experimental protocol was obtained from the University of Michigan Animal Care and Use Committee.
  • Stock nanoemulsion W 20 5GBA 2 ED was obtained from NanoBio Corporation (Ann Arbor, Mich.). This nanoemulsion was manufactured by emulsification of super-refined soybean oil and water with surfactants and alcohol as described herein. The resultant droplets had a mean particle diameter of 350 nm.
  • the experimental solution was made by diluting 1 mL of the 60% stock formulation with 4.88 mL sterile saline and adding 120 ⁇ L of 1 M ethylenediaminetetraacetic acid (EDTA) giving a final concentration of 10% W 20 5GBA 2 ED and 20 mM EDTA.
  • EDTA ethylenediaminetetraacetic acid
  • a placebo nanoemulsion (W 20 5GBA 2 ED placebo) compound was manufactured in the same manner as W 20 5GBA 2 ED, but benzalkonium chloride was omitted from the formulation.
  • 5% Sulfamylon (UDL Laboratories, Inc., Rockford, Ill.) solution was formulated by mixing 50 g of mafenide acetate powder in 1 L of 0.9% sterile saline. The control reagent used was 0.9% sterile saline.
  • Pseudomonas aeruginosa isolated from a human burn patient was previously provided by the Department of Pathology at the University of Michigan. This bacterial isolate is sensitive to the topical agent Silvadene and Sulfamylon.
  • a bacterial inoculum was prepared by thawing an aliquot (0.5 mL, stored in 50% skim milk at ⁇ 80° C.) in 40 mL of Trypticase soy broth (Becton Dickinson, Franklin Lakes, N.J.) and grown overnight at 37° C. with constant shaking at 275 rpm.
  • a sample of the resulting stationary-phase culture was transferred to 35 mL of fresh Trypticase soy broth and incubated for 2.5 hours to reach the log-phase.
  • This subculture was transferred to a 50 mL conical polystyrene tube and centrifuged for 10 minutes at 4° C. and 880 g.
  • the bacterial pellet was washed with 0.9% sterile saline, and resuspended in 10 mL of ice-cold saline.
  • the optical density of the suspension was measured at 620 nm and bacterial concentration (colony forming units (CFU)/mL) calculated using the formula OD 620 ⁇ 2.5 ⁇ 10 8 .
  • the bacterial suspension was diluted with 0.9% sterile saline to a final concentration of 1 ⁇ 10 6 CFU per 100 ⁇ L.
  • Eight hours following burn injury animals were anesthetized with inhaled isoflurane.
  • the rats then underwent topical application of 1 ⁇ 10 6 CFUs of log-phase Pseudomonas aeruginosa in 100 ⁇ L of sterile saline pipetted onto a piece of TELFA in a uniform fashion followed by coverage with a TEGADERM occlusive dressing.
  • Tissue harvest Thirty-two hours after thermal injury the animals were sacrificed and skin tissue samples harvested using sterile technique. Skin samples were used immediately or frozen in liquid nitrogen.
  • ELISA Dermal cytokine analysis
  • the assay was carried out in 96-well microplates (Immunoplate Maxisorb, Nunc, Neptune, N.J.) according to the kit instructions and samples were read using a microplate reader (Biotek Instruments, Winooski, Vt.) at 450 nm with a wavelength correction of 540 nm. Cytokine concentrations were determined using the plate reader software and a 7-point standard curve. Results were adjusted for previous dilution and expressed as pg/mL.
  • Detection of neutrophil sequestration Myeloperoxidase assay. 100 mg of skin tissue was mechanically homogenized in 1 mL ice cold potassium phosphate buffer consisting of 115 mM monobasic potassium phosphate (Sigma Aldrich, Milwaukee, Wis.). Homogenates were centrifuged at 3000 g for 10 min at 4° C., the supernatants were removed and the pellets were re-suspended in 1 mL C-TAB buffer consisting of dibasic potassium phosphate, cetyltrimethylammonium bromide, and acetic acid (Sigma Aldrich, Milwaukee, Wis.).
  • the suspensions were sonicated (Branson Sonifier 250, Danbury, Conn.) on ice for 40 seconds. Homogenates were centrifuged at 3000 g for 10 min at 4° C. and the supernatant collected. Supernatants were incubated in 60° C. water bath for 2 hours (Shaker Bath, 2568; Form a Scientific, Marietta, Ohio). Samples were stored at ⁇ 80° C. until needed or assayed immediately.
  • TUNEL assay Detection of hair follicle cell apoptosis. Animals were anesthetized and underwent creation of a 20% partial thickness scald burn wound or sham injury.
  • Treatment groups consisted of sham, burn+saline, burn+placebo, and burn+W 20 5GBA 2 ED. Treatment and dressing changes were performed at 0 and 8 hours post-burn. No bacterial infection was created in this experiment. Full-thickness skin samples were taken from three locations across the entire burn wound at 12, and 24 hours post thermal injury for determination of hair follicle cell apoptosis. There were four animals per treatment group per time sample.
  • Apoptosis was detected in situ with fluorescein based labeling of DNA strand breaks using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay (ApopTag, CHEMICON International, Inc, Temecula, Calif.).
  • TUNEL terminal deoxynucleotidyl transferase dUTP nick end labeling
  • ApopTag CHEMICON International, Inc, Temecula, Calif.
  • the three fresh skin samples for each animal were placed in disposable vinyl cryomolds filled with optimal cutting temperature compound (Sakura Finetek, U.S.A., Inc., Torrance, Calif.), and frozen at ⁇ 80° C. until ready for use. Frozen embedded skin specimens were cut into 4-mm-thick serial sections in a cryostat and collected on Superfrost Plus glass slides (Fisher Scientific, Pittsburgh, Pa.).
  • Sections were fixed in 1% paraformaldehyde in PBS overnight at 4° C. for indirect immunofluorescence according to the manufacturer's instructions. After 2 washes with PBS, sections were post-fixed in a 2:1 solution of ethanol:acetic acid for 5 minutes at ⁇ 20° C. Following 2 additional washes with PBS, sections were incubated in equilibrium buffer for 5 minutes. Sections were then incubated with terminal deoxynucleotidyl transferase enzyme in a humidified chamber for 1 h at 37° C. The reaction was terminated by rinsing the sections in a stop/wash buffer.
  • Sections were incubated in a humidified chamber at room temperature with antidigoxigenin fluorescein (fluorescein isothiocyanate, FITC) for 30 min and rinsed 3 times in PBS. After aspiration, sections were washed once with water and coverslips applied using ProLong Gold Antifade (Molecular Probes, Inc, Eugene, Oreg.), which included 4′ 6-diamidino-2-phenylindole dihydrochloride (DAPI) counterstaining.
  • FITC fluorescein isothiocyanate
  • the TUNEL assay slides were blinded to groups, and under the microscope appropriate hair follicle cells in a randomly chosen high-power field were identified. Appropriate hair follicles for analysis were those sectioned in the mid-sagittal or mid-coronal plane. A total of 3-6 hair follicles were selected from among the three skin samples present on a slide. Fluorescent-labeled TUNEL slides were captured digitally at identical time post-labeling to control fading of fluorescence using an Olympus BX-51 fluorescence microscope at fixed image capture settings and 40 ⁇ magnification. Each hair follicle was selected and first digitally captured by visualizing counterstained nuclei present using the DAPI excitation/emission channel.
  • the excitation/filter channel was changed to visualize the fluorescein-labeled TUNEL-positive cells, and images again digitally captured.
  • a region of interest ROI was digitally defined, set to include only hair follicle cells and exclude bright fluorescing hair shafts and surrounding cells (NIH Image J software, NIH, Bethesda, Md.). Fluorescence of TUNEL-positive cells was quantified, normalized to ROI size, and expressed as pixels/area fraction, controlling for differences in ROI size.
  • a positive result is generally considered to be growth of organisms at greater than 1 ⁇ 10 5 CFUs per g of tissue (See, e.g., Neal et al., J Burn Care Rehabil 2:35-39, 1981; Taddonio et al., Burns Incl Therm Inj 14(3):180-184, 1988; Uppal et al., Burns 33(4):460-463, 2007).
  • 29 of 32 animals in the control group exhibited evidence of a positive quantitative wound culture and only 3 of 23 animals in the nanoemulsion group demonstrated proof of this level of wound infection (91% vs. 13%, p ⁇ 0.0001, See Table 24, below).
  • the Sulfamylon treated animals also demonstrated a significant reduction in the median wound bacterial level when compared to the saline controls (3 ⁇ 10 4 vs. 4.4 ⁇ 10 6 , p ⁇ 0.05).
  • Treatment with nanoemulsion or Sulfamylon produced a similar reduction in the level of Pseudomonas cultured from the burn wound when compared to the saline treated animals.
  • there was no statistically significant difference between the placebo and Sulfamylon groups whereas there was a difference for the W 20 5GBA 2 ED group compared to the W 20 5GBA 2 ED placebo.
  • Scald injury resulting in a partial thickness burn produced differences in dermal levels of IL-1 ⁇ and cytokine-induced neutrophil chemoattractant-3 (CINC-3) within skin homogenates obtained 32 hours post-injury compared to sham injured animals (See FIGS. 22A and E).
  • Treatment with W 20 5GBA 2 ED at 16 and 24 hours post-burn reduced the dermal level of these two inflammatory mediators back down to the baseline (sham) in the absence of bacterial infection.
  • a difference in neutrophil sequestration as measured by myeloperoxidase assay was not observed despite the rise in the rat CXC chemokine CINC-3 within burned skin.
  • Nanoemulsion Treatment Following Burn Wound Infection with P. aeruginosa Attenuates Dermal Cytokine Levels and Results in Reduced Neutrophil Sequestration
  • treatment with either W 20 5GBA 2 ED or Sulfamylon reduced the level of myeloperoxidase found in the infected burn wound at 32 hours post-injury.
  • treatment with a nanoemulsion and/or antimicrobial reduces the level of neutrophil sequestration into a partial thickness burn wound.
  • Burn injury caused a rise in the level of the anti-inflammatory cytokine TGF- ⁇ , but not IL-10 when compared to the sham injured animals (See FIG. 24 ).
  • W 20 5GBA 2 ED treatment reduced the amount of TGF- ⁇ present in the infected burn wound as compared to the level found in the burn wound alone. Accordingly, in some embodiments, the present invention provides that W 20 5GBA 2 ED can be utilized to reduce acute burn wound dermal inflammation, and in further embodiments, can be utilized to reduce the eventual immunosuppression created by thermal injury.
  • the keratin layers of the epidermis are separating and some of the keratin has been lost.
  • the changes in this microphotograph are substantially less extensive than those seen in FIG. 25A .
  • Burn wounds are associated with significant levels of capillary leak. This can lead to depletion of the intravascular volume and a need for large amounts of intravenous crystalloid fluid administration.
  • an Evans blue assay was utilized to measure vascular permeability. Quantitative measurement of the amount of this dye leaching out of the blood stream and into the skin tissue revealed that the nanoemulsion treated animals had less evidence of post-burn capillary leak and tissue edema than the saline treated controls (See FIG. 26 ).
  • Dermal apoptosis occurs in the hair follicle cells following thermal injury.
  • TUNEL assay the burn wounds treated with saline showed evidence of intense FITC-TUNEL positive cells which appear green (See FIG. 27 ).
  • the DAPI nuclear stain allows identification of coronal or sagittally sectioned hair follicles with the cells staining blue.
  • FITC-TUNEL positive cells appear green and are representative of apoptotic cells.
  • the apoptotic hair follicle cells are evident in slides from the burn+saline animals and these changes are diminished in the burn+W 20 5GBA 2 ED treated animals.
  • Counting the pixels of TUNEL positive cells within a hair follicle region of interest allowed quantification of the reduction in hair follicle cell apoptosis by treatment with topical nanoemulsion (See FIG. 28 ).
  • the saline treated control animals had an increased amount of TUNEL positive cells when compared to the sham burn animals.
  • Both the W 20 5GBA 2 ED and placebo treatment resulted in a decrease in hair follicle cell apoptosis following partial thickness burn injury in tissue harvested 12 hours following thermal injury. This difference was not evident in the dermal skin sampled at 24 hours post-burn.
  • the present invention provides a method of reducing apoptotic cell death in hair follicles in the early post-burn period comprising administering a nanoemulsion (e.g., W 20 5GBA 2 ED) to a burn wound during an early post-burn period.
  • a nanoemulsion e.g., W 20 5GBA 2 ED
  • Burkholderia multivorans ATCC 17616 was used for cross-strain neutralization assays. Both K56-2 and ATCC 17616 strains were stored at ⁇ 80° C. in Luria-Bertani broth with 15% glycerol and recovered from frozen stock on brain-heart infusion agar overnight at 37° C.
  • OMP cenocepacia Outer Membrane Protein
  • the sonicated lysates were then centrifuged twice for 10 minutes at 6000 ⁇ g (Beckman Optima XL-100 k ultracentrifuge, 25° C.) with the supernatant retained after each spin. Following the second centrifugation, the supernatant was centrifuged at 100,000 ⁇ g for 1 hour at 4° C. The resulting pellet was resuspended in endotoxin-free PBS.
  • the rough OMP preparation was purified (endotoxin-depleted OMP) with an endotoxin-removal column (Pierce) according to the manufacturer's instructions. The flow-through fractions were stored at ⁇ 80° C. until used.
  • OMP Analysis The protein contained within the OMP preparation was quantified using BCA Assay (Pierce). Western blotting and silver staining were performed according to established protocol as described previously (See, e.g., Makidon et al., 2008, PLoS ONE 3, e2954). To quantify endotoxin contaminate, OMP was analyzed using the LAL Kinetic-QCL (Lonza). Endotoxin was detected at 1.93 endotoxin units/mg in the endotoxin-depleted OMP preparation. DNA contaminant removal was verified by agarose-EtBr gel electrophoresis and imaged on a UV table (See FIG. 29A ). Oligonucleotides contaminants were not detected in either the crude OMP or the endotoxin-depleted OMP preparations.
  • the protein sample was separated by SDS-PAGE (See FIG. 29B ).
  • the protein band immunostained with the highest intensity ( ⁇ 17 KDa) on western blot See FIG. 29C ) was excised and subjected to in-gel digestion.
  • In-gel digestion was performed at the Protein Structure Facility at the University of Michigan according to procedures by Rosenfeld et al. and Hellman et al. See, e.g., Hellman et al., 1995, Analytical Biochemistry 224, 451-455; Rosenfeld et al., 1992, Analytical Biochemistry 203, 173-179).
  • a gel slice containing 5- ⁇ mol bovine serum albumin (BSA) was analyzed in parallel as a positive control.
  • On-line capillary HPLC was performed using a Waters UPLC system with an Atlantis C18 column (Waters, 100-mm inner diameter, 100-mm length, 3-mm particle size). Digests were desalted using an online trapping column (Waters, Symmetry C18, 180-mm inner diameter, 20-mm length, 5-mm particle size) before being loaded onto the Atlantis column.
  • a data-dependent tandem mass spectrometry approach was utilized to identify peptides in which a full scan spectrum (survey scan) was used, followed by collision-induced dissociation (CID) mass spectra of all ions in the survey scan with a peak intensity rising above 20 counts per second.
  • the survey scan was acquired in V+mode over a mass range of 50-2000 Da with lockmass correction (Gu-Fibrinopeptide B, (M+H) 2+ : 785.8426) and charge state peak selection (2+, 3+ and 4+).
  • the MS/MS scans were acquired for 5 seconds with collision energy control by charge state recognition.
  • Tandem mass spectra were acquired using Mass Lynx software (Version 4.1). Raw data files were first processed for lockmass correction, noise reduction, centering, and deisotoping using the Protein Lynx Global Server software Ver. 4.25 (Waters Inc.). The generated peaklist files containing the fragment mass spectra were subjected to database searches using the ProteinLynxGlobal Server and Mascot (Matrix Science Inc., Boston, Mass.) search engines and Swiss Prot and NCBI databases, as well as the Burkholderia cenocepacia database downloaded from the Sanger Institute web site.
  • Nanoemulsion-based OMP (OMP-NE) vaccine Preparation of Nanoemulsion-based OMP (OMP-NE) vaccine.
  • Nanoemulsion W 80 5EC: 5 vol. % of TWEEN 80, 8 vol. % of ethanol, 1 vol. % of CPC, 64 vol. % of soybean oil) and 22 vol. % of DiH 2 O
  • OMP-NE formulations were prepared by vigorously mixing the endotoxin-depleted OMP preparation (See FIG. 29B , Lane F) with concentrated NE, using PBS as the diluent.
  • OMP-NEs were formulated to contain either 0.25 ⁇ g/mL or 0.75 ⁇ g/ ⁇ L OMPs mixed in 20% (v/v) NE.
  • the anesthetized animals were held in a supine position and 20 ⁇ L (10 ⁇ L/nare) of OMP-NE vaccine was administered slowly to the nares using a micropipette tip.
  • Mice were immunized with either 15 ⁇ g OMP with NE, 15 ⁇ g OMP in PBS, 5 ⁇ g OMP with NE, or 5 ⁇ g OMP in PBS.
  • Phlebotomy, bronchoalveolar lavage, and splenocyte collection Blood was collected from the saphenous vein every 21 days throughout the duration of the study. The terminal sample was obtained by cardiac puncture immediately following euthanasia. Whole blood samples were separated by centrifugation at 3500 rpm (15 minutes) following coagulation. The serum samples were stored at ⁇ 20° C. until analyzed. Bronchoalveolar lavage (BAL) fluid was obtained from the mice immediately following euthanasia as described (See Makidon et al., 2008, PLoS ONE 3, e2954). In vitro measurement of the cytokine response was determined using the spleens of the vaccinated mice.
  • Spleens were mechanically disrupted to obtain single-cell splenocyte suspension in PBS.
  • Red blood cells were lysed with ACK buffer (150 mM NH4Cl, 10 mM KHCO3, 0.1 mM Na2EDTA) and removed by washing the cell suspension twice in PBS.
  • the splenocytes were then resuspended in RPMI 1640 medium supplemented with 2% FBS, 200 nM L-glutamine, and penicillin/streptomycin (100 U/mL and 100 mg/mL).
  • Enzyme-linked immunosorbent assay Serum IgG and mucosal secretory IgA (sIgA) were determined by ELISA against endotoxin-depleted OMP.
  • the OMP was prepared at 15 ⁇ g/mL in coating buffer (Sigma) and 100 ⁇ L was applied per well to Polysorb plates (Nunc) for a 4° C. overnight incubation.
  • Serum was serially diluted in 0.1% BSA/PBS. Either diluted serum or undiluted BAL was then added to the plates for an overnight incubation at 4° C.
  • H&L alkaline phosphatase conjugated anti-mouse IgG
  • IgG1, IgG2a, IgG2b, sIgA a chain-specific antibody
  • BSA/PBS alkaline phosphatase conjugated anti-mouse IgG
  • IgG1, IgG2a, IgG2b, sIgA a chain-specific antibody
  • serum lipoprotein-specific and lipopolysaccharide-specific IgG antibodies in serum were measured using ELISA. Briefly, 96-well Polysorb assay plates (Nunc) were coated with 0.05 ⁇ g/well of crude OMP preparation, endotoxin-depleted OMP, or LPS (List Biological Laboratories). Serum was collected from mice immunized with OMP+NE 8 weeks following prime vaccination. The serum was serially diluted in 0.1% BSA/PBS and then added to the coated wells. The ELISA procedure was performed as described supra.
  • an enzymatic protein digest was performed using 250 ⁇ g of PCR-grade recombinant proteinase K (Roche) in 100 ⁇ l of either the crude OMP preparation or the endotoxin-depleted OMP preparation. Proteinase K was incubated with the OMP preparations for 16 hours at room temperature. The protein digestion was verified using a silver-stained SDS-PAGE. LPS and OMP-protein specific epitopes were evaluated using western blot analysis probing with serum from mice vaccinated with either OMP-NE or OMP in PBS, or probing with an anti- Pseudomonas mallei LPS antibody (GeneTex).
  • cytokine expression Freshly isolated mouse murine splenocytes were seeded at 4 ⁇ 10 6 cells/mL (RPMI 1640, 2% FBS) onto a 24-well plate and incubated for 72 hours with OMPs (15 ⁇ g/mL) or PHA-P mitogen (2 ⁇ g/mL) as a control. Cell culture supernatants were collected and analyzed for the presence of cytokines using LUMINEX multi-analyte profiling beads (LUMINEX Corporation, Austin, Tex.), according to the manufacturer's instructions.
  • LUMINEX multi-analyte profiling beads LUMINEX Corporation, Austin, Tex.
  • B. cenocepacia strain K56-2 and B. multivorans strain ATCC 17616 antibody-complement neutralization activity was assayed using serum harvested from mice 6 weeks following primary i.n. vaccination. Twenty-five ⁇ L of serum (serially diluted 1 ⁇ , 2 ⁇ , 3 ⁇ , 4 ⁇ , or 5 ⁇ ) was added to 25 ⁇ L of a solution containing 1 ⁇ 10 3 K56-2 or B. multivorans supplementing MH medium in microculture plates (Nunc). The plates were incubated at 37° C. for 48 hours. Controls included 25 ⁇ L serum with 25 ⁇ L of sterile 1 ⁇ PBS and 50 ⁇ L of MH medium containing 1 ⁇ 10 3 bacteria.
  • BCSA Burkholderia selective agar
  • mice were maintained for a period of 6 days. Prior to the challenge study, the mice were non-surgically implanted with programmable temperature transponders (IPTT-3000, Bio Medic Data Systems, Inc.) for non-invasive subcutaneous temperature measurement with a handheld portable scanner (DAS-6002, Bio Medic Data Systems, Inc.). In-life analysis included monitoring the clinical status of each animal by evaluating the following metrics: body weight, body temperature, food consumption, and activity. Immediately following euthanasia, pulmonary tissues and the spleen were collected in sterile conditions and each organ was placed into individual containers containing 1 mL of 1 ⁇ PBS (Mediatec Inc). The tissue was then homogenized using a Tissue TEAROR (Biospec Products Inc.) for 50 seconds on ice.
  • IPTT-3000 programmable temperature transponders
  • DAS-6002 Bio Medic Data Systems, Inc.
  • Immunoreactive OMP proteins were identified via silver stain (See FIG. 29B ) and western blot (See FIGS. 29C and D). Major reactive bands were documented at 62, 45, 17, and 10 KDa in both cases; however, the intensity of the bands was much higher with serum from mice immunized with the NE-based vaccine (See FIG. 29C ) as compared to the blot probed with an equivalent serum dilution from mice immunized with the OMP in PBS preparation (See FIG. 29D ).
  • the band with the highest intensity was located at ⁇ 17 KDa (See FIG. 29C ).
  • the 17 KDa band was isolated from the gel and identified by MALDI-TOF analysis.
  • the protein was identified as Burkholderia cepacia outer membrane lipoprotein A (OmpA) with a MW of 16.396 KDa (See, e.g., Ortega et al., 2005 J Bacteriol 187, 1324-1333; Plesa et al., 2004, J Med Microbiol 53, 389-398).
  • Sequence analysis using National Center for Biotechnology Information (NCBI) protein BLAST confirmed that these proteins are present in other closely related Burkholderia species (See Table 25, below).
  • a high degree of protein homology (87.9% sequence homology) was identified in numerous strains of Burkholderia and Ralstonia organisms and highly conserved amino acids from the OmpA family residues are present in the 17 KDa protein sequence (See Table 25).
  • the present invention identified and provides immunoreactive OMP proteins from B. cepacia as well as proteins from related strains of Burkholderia species displaying a high degree of homology to the identified immunoreactive OMP proteins from B.
  • cepacia e.g., that find use in immunogenic compositions comprising a nanoemulsion (e.g., that are administered to a subject in order generate a specific immune response in the subject toward the immunogenic composition comprising nanoemulsion and immunoreactive OMP proteins or homologues)).
  • the humoral immune responses against the OMP induced by nasal vaccine with OMP were characterized in vivo in the CD-1 mice.
  • Intranasal vaccination with either 5 ⁇ g or 15 ⁇ g OMP preparations+NE (OMP-NE) resulted in high serum titers of OMP-specific IgG antibodies of 2.8 ⁇ 10 5 and 5.1 ⁇ 10 5 at 6 weeks, respectively, following primary vaccination (See FIG. 30A ).
  • OMPs without adjuvant were immunogenic and resulted in serum anti-OMP IgG titers of 1.9 ⁇ 10 4 and 3.8 ⁇ 10 4 six weeks following primary i.n. immunization.
  • mice immunized with OMP in PBS did not demonstrate a significant boost effect following the second vaccination (See FIG. 30A ).
  • the OMP-NE composition's ability to elicit specific antibody production in bronchiolar secretions was evaluated. Mucosal antibody production may be important for protection against Burkholderia colonization since secretory antibodies are thought to be critical effectors in protection against mucosal respiratory pathogens (See, e.g., Nelson et al., 1993, J Med Microbiol 39, 39-47). Mucosal immune responses were evaluated in bronchoalveolar lavage (BAL) fluid of animals immunized with 5 ⁇ g OMP-NE or 5 ⁇ g OMP in PBS. Comparable levels of anti-OMP antibodies in BALs were detected in mice immunized either with OMP-NE or OMP without NE adjuvant (See FIG. 30B ).
  • BAL bronchoalveolar lavage
  • OMP-NE Immunization Yields a Balanced Th1/Th2 Cellular Response
  • the analysis of the serum IgG subclass was performed to determine the T helper-type bias of the cellular response.
  • immunization with OMP in PBS produced similar levels of IgG1 and IgG2b antibodies (See FIG. 31A ).
  • Th2 cytokines IL-4, IL-5, IL-6, and IL-10 were minimally induced ( ⁇ 5.22-fold increase) in both OMP-NE and OMP immunized mice in equivalent amounts.
  • Antibody Response is Directed Toward OMP Lipoprotein and LPS
  • the present invention provides that nasal immunization with OMP-NE enhances the production of antibodies specific against OMP proteins (See FIG. 32 ).
  • B. cenocepacia neutralizing antibodies were determined using a colony reduction assay.
  • the mice intranasally vaccinated with 5 ⁇ g or 15 ⁇ g OMP-NE produced serum-neutralizing antibodies that inhibited 92.5% or 98.3% of B. cenocepacia colonies, respectively, as compared to serum from non-vaccinated mice (See FIG. 33 ).
  • the mice vaccinated with OMP lacking adjuvant also demonstrated a relatively high level of inhibition, resulting in 33.3% and 46.7% (for 5 ⁇ g and 15 ⁇ g OMP-PBS, respectively) cfu reduction.
  • the statistical analysis showed that B. cenocepacia growth was significantly inhibited by serum derived from OMP-NE as compared to serum from those who were immunized with the OMP preparation alone (See FIG. 33 ).
  • mice vaccinated with 15 ⁇ g OMP-NE was also incubated with B. multivorans .
  • the B. multivorans was selected because it is the most common isolate cultured from CF patients infected with Bcc organisms (See, e.g., Baldwin et al., 2008, J Clin Microbiol 46, 290-295).
  • the protective effect of intranasal immunization was further tested in vivo in a lung infection model.
  • the clearance of B. cenocepacia from pulmonary tissue was evaluated 6 days following intra-tracheal inoculation in mice that were intranasally immunized with the OMP-NE or the OMP without an adjuvant.
  • the average pulmonary bacterial load was 22.5 ⁇ 26.2 cfu in mice vaccinated with 15 ⁇ g OMP-NE, compared to 1.28 ⁇ 10 6 ⁇ 3.36 ⁇ 10 6 cfu per lung in non-vaccinated mice, representing a greater than 5-log reduction in the bacterial load.
  • Splenic colonization following pulmonary inoculation with B. cenocepacia was evaluated as a means of assessing sepsis (See FIG. 34B ).
  • B. cenocepacia infection resulted in greater loss of thermoregulation by day 6 in the non-vaccinated mice than in the mice immunized with OMP-NE or OMP without the adjuvant.
  • the present invention provides that immunization with OMP-NE resulted in significantly decreased systemic disease following infection with B. cenocepacia .

Abstract

The present invention relates to therapeutic nanoemulsion compositions and to methods of utilizing the same. In particular, nanoemulsion compositions are described herein that find use in the treatment and/or prevention of infection (e.g., respiratory infection (e.g., associated with cystic fibrosis)), in burn wound management, and in immunogenic compositions (e.g., comprising a Burkholderia antigen) that generate an effective immune response (e.g., against a bacterial species of the genus Burkholderia) in a subject administered the immunogenic composition. Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine), industrial, and research applications.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 60/100,559 filed Sep. 26, 2008, the entire disclosure of which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to therapeutic nanoemulsion compositions and to methods of utilizing the same. In particular, nanoemulsion compositions are described herein that find use in the treatment and/or prevention of infection (e.g., respiratory infection (e.g., associated with cystic fibrosis)), in burn wound management, and in immunogenic compositions (e.g., comprising a Burkholderia antigen) that generate an effective immune response (e.g., against a bacterial species of the genus Burkholderia) in a subject administered the immunogenic composition. Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine), industrial, and research applications.
  • BACKGROUND OF THE INVENTION
  • Bacterial infection caused by opportunistic and/or pathogenic bacteria (e.g., Burkholderia cepacia, Staphylococcus aureus and Pseudomonas aeruginosa) is a major problem in both the developed and undeveloped portions of the world. For example, certain types of individuals are prone to respiratory infection (e.g., by bacteria (e.g., opportunistic bacteria), viruses, fungi and/or parasites) including the immunocompromised, elderly, cancer chemotherapy patients, individuals suffering from asthma, individual suffering from genetically inherited disease (e.g., cystic fibrosis) and virally infected individuals (e.g., infected with influenza virus, respiratory syncytial virus (RSV), adenovirus and/or human immunodeficiency virus). Similarly, wounds (e.g., burn wounds) present on a subject provide an ideal location for bacterial growth and survival.
  • As the use of conventional pharmaceutical antibiotics has increased for medical, veterinary and agricultural purposes, there has been a concurrent emergence of antibiotic-resistant strains of pathogenic bacteria.
  • A need exists to develop alternative strategies of antibacterial treatment. For example, there exists a need for new compositions and methods of treating or preventing bacterial infection (e.g., bacteremia) caused by strains of bacteria unsusceptible to current forms of antibacterial treatments.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to a method of treating or preventing a respiratory infection in a subject having Cystic fibrosis (CF). The method comprises administering a nanoemulsion to the subject, wherein the subject is susceptible to or has an infection by one or more gram-positive or gram-negative bacterial species. The nanoemulsion comprises water, at least one organic solvent, at least one surfactant, and at least one oil. In addition, the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm. In one embodiment of the invention, the respiratory infection is associated with a bacterial biofilm present in the lungs of the subject.
  • In another embodiment of the invention, described is a method of treating or preventing an infection in a subject having a burn wound. The method comprises administering a nanoemulsion to the subject, wherein the subject is susceptible to or has an infection by one or more gram-positive or gram-negative bacterial species. The nanoemulsion comprises water, at least one organic solvent, at least one surfactant, and at least one oil. In addition, the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • For methods directed to subjects having a or susceptible to a gram-negative or gram-positive bacterial infection, the bacteria can be any known gram-negative or gram-positive bacteria. In another embodiment of the invention, the bacterial species is selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp. The bacterial species can also be selected from the group consisting of Pseudomonas aeruginosa, B. cenocepacia, A. baumannii, Stenotrophomonas maltophilia, Staphylococcus aureus, H. influenzae, E. coli, K. pneumoniae, and Proteus mirabilis.
  • In yet another embodiment of the invention, described is a method of treating or preventing an Haemophilus influenzae infection in a non-CF subject. The method comprises administering a nanoemulsion to the subject having or at risk of having a Haemophilus influenzae infection. The nanoemulsion comprises water, at least one organic solvent, at least one surfactant, and at least one oil. In addition, the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
  • All methods described herein may further comprise administering one or more antibiotics either before, during, or after administration of the nanoemulsion. In one embodiment, the administration of a nanoemulsion and at least one antibiotic is synergistic. The “synergy” may be as defined by a fractional inhibitory concentration (FIC) index, a fractional bactericidal concentration (FBC) index, or a combination thereof. Methods comprising administering an antibiotic encompass the use of any antibiotic. Exemplary antibiotics include, but are not limited to, a polymyxin antibiotic (e.g. colistin) or any aminoglycoside (e.g. tobramycin). Preferably, in the methods of the invention utilizing an antibiotic, the nanoemulsion does not exhibit any antagonism with the antibiotic.
  • For all methods described herein, preferably the nanoemulsion exhibits minimal or no toxicity or side effects. In addition, preferably the nanoemulsion does not exhibit resistance. In another embodiment applicable to all methods described herein, the minimum inhibitory concentration (MIC), the minimum bactericidal concentration (MBC), or a combination thereof for the nanoemulsion demonstrate bacteriostatic or bactericidal activity for the nanoemulsion.
  • For all methods described herein, one or more bacterial species may exhibit resistance against one or more antibiotics. For example, the bacterial species can be methicillin-resistant Staphylococcus aureus (MRSA).
  • In one embodiment of the methods of the invention, the nanoemulsion is not absorbed systemically in the human subject, or very little of the nanoemulsion is absorbed systemically in the human subject.
  • In the methods of the invention, the nanoemulsion can be delivered using any pharmaceutically acceptable means. For example, the nanoemulsion can be delivered via inhalation, topically, topically to a mucosal surface, via nebulization, or via any combination thereof.
  • The invention also encompasses compositions useful in the methods of the invention. Exemplary compositions include, but are not limited to, nanoemulsions comprising: (a) water; (b) ethanol or glycerol; (c) cetylpyridinium chloride (CPC), or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824); (d) soybean oil; and (e) Poloxamer 407, Tween 80, or Tween 20. In a further embodiment, the nanoemulsion can additionally comprise EDTA.
  • The foregoing general description and following brief description of the drawings and the detailed description are exemplary and explanatory and are intended to provide further explanation of the invention as claimed. Other objects, advantages, and novel features will be readily apparent to those skilled in the art from the following detailed description of the invention.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of necessary fee.
  • FIG. 1 shows killing of Burkholderia cepacia in PBS by 20% Nanemulsion (NE) alone or with 20 mM EDTA in 10 minutes.
  • FIG. 2 shows killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 20 minutes.
  • FIG. 3 shows killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 40 minutes.
  • FIG. 4 shows (A) killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 40 minutes; (B) killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 60 minutes; and (C) killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA in 60 minutes.
  • FIG. 5 shows killing of B. cepacia in hypertonic saline (6% NaCl) at 15 and 30 Minutes by NE alone and NE with EDTA.
  • FIG. 6 shows killing of B. cepacia and P. aeruginosa in 7% NaCl within 15 min.
  • FIG. 7 shows killing of B. cepacia and P. aeruginosa (Mixed-culture) in 7% NaCl within 15 minutes.
  • FIG. 8 shows a microtiter serial dilution MIC assay. Numbers above the plate indicate the concentration (μg/ml CPC) of P4075EC added to each column of wells. The bacterial species added to wells in each row is indicated on the left. The MICs for these strains (top to bottom) are ≦15.6 μg/ml 125 μg/ml, and 31.2 μg/ml.
  • FIG. 9 shows distribution of P4075EC MICs for 150 Burkholderia and non-Burkholderia strains
  • FIG. 10 shows a time-kill study of P4075EC activity and concentration-dependent inhibition of P4075EC activity by CF sputum. (A) 105 CFU/ml of B. multivorans ATCC 17616 were exposed to various concentrations of P4075EC around the MIC (62.5 μg/ml) and viable bacterial counts were determined at the times indicated. (B) 105 CFU/ml of B. multivorans ATCC 17616 were exposed to various concentrations of P4075EC in the presence of three concentrations of CF sputum for 24 hr before viable bacterial counts were determined. Numbers and symbols in inset boxes indicate concentrations of P4075EC (μg/ml CPC) used.
  • FIG. 11 shows in vitro activity of P4075EC.
  • FIG. 12 shows in vitro activity of P4075EC against biofilm bacteria and in the presence of CF sputum.
  • FIG. 13 shows influence of P4075EC on B. cepacia, P. aeruginosa and E. coli individual biofilms.
  • FIG. 14 shows influence of P4075EC on mixed biofilms.
  • FIG. 15 shows influence of P4075EC on mixed biofilms.
  • FIG. 16 shows influence of P4075EC on escape colonies of B. cepacia biofilms.
  • FIG. 17 shows serial, two-fold dilutions of P4075EC in one embodiment of the invention.
  • FIG. 18 shows age specific prevalence of respiratory infections in CF patients.
  • FIG. 19A shows a Fractional Inhibitory Concentration Index;
  • FIG. 19B shows a graph demonstrating the relationship between antagonism, indifference, and synergy for Antimicrobial A (micrograms/mL) and Antimicrobial B (micrograms/mL);
  • FIGS. 20A-D show figures of electron microscopy. FIG. 2A shows 0 min, no treatment control; FIG. 2B shows 10 minutes after treatment; FIG. 2C shows 20 minutes after treatment; and FIG. 2D shows 30 minutes after treatment.
  • FIG. 21 shows topical application of nanoemulsion reduces P. aeruginosa growth in burn wounds. Male Sprague-Dawley rats received partial thickness burn wounds. At 8 hours post-injury, animals were inoculated with 106 CFU P. aeruginosa. At 16 and 24 hours post-burn the animals were treated with topical saline (Control), placebo (W205GBA2ED without benzalkonium chloride), W205GBA2ED (nanoemulsion), or Sulfamylon (mafenide acetate). At 32 hours, animals were sacrificed, skin samples obtained and homogenized, plated, and CFUs counted. The scatter plot represents cultured CFUs for each individual animal. The median value for each group is plotted as a horizontal line. There was minimal pathogen growth in 12 out of 23 of the NB-201 treated animals. A majority of the control (29/32) and placebo (9/12) animals with burn injury had evidence of wound infection based on a positive quantitative wound culture with significantly more bacteria present in the wound than those animals treated with W205GBA2ED. p<0.0001, Kruskal-Wallis test, p<0.05 for saline vs. W205GBA2ED, placebo vs. W205GBA2ED, and saline vs. Sulfamylon, Dunn's multiple comparison test.
  • FIG. 22 shows nanoemulsion treatment following burn injury attenuates dermal proinflammatory cytokine expression. Groups were sham, burn, and burn with W205GBA2ED treatment (n=8-10 per group). A) IL-43 (p=0.02, 1-way ANOVA), B) IL-6 (p=0.8), C) TNF-α (p=0.5), D) CINC-1 (p=0.04), E) CINC-3 (p=0.005), and F) Myeloperoxidase (p=0.07). Burn injured animals treated with nanoemulsion had decreased levels of IL-1β (1773±516 vs. 5625±1743 pg/mL) and CINC-3 (225±66 vs. 1589±527 pg/mL) when compared to untreated partial thickness burned animals. *p<0.05, t-test or Tukey's multiple comparison test.
  • FIG. 23 shows nanoemulsion treatment following burn wound infection with P. aeruginosa attenuates dermal proinflammatory cytokine expression. All animals received burn injury and groups were saline, placebo, W205GBA2ED, and Sulfamylon (n=10-30 per group). A) IL-1β (p=0.001, 1-way ANOVA), B) IL-6 (p=0.07), C) TNF-α (p=0.3), D) CINC-1 (p=0.8), E) CINC-3 (p=0.4), and F) Myeloperoxidase (p=0.0001). Burn wound infected animals treated with nanoemulsion had decreased levels of IL-1β (1007±157 vs. 3054±499 pg/mL) and IL-6 (244±51 vs. 485±73 pg/mL). The nanoemulsion and Sulfamylon treatment groups demonstrated reduced dermal neutrophil sequestration when compared to saline and placebo as evidenced on myeloperoxidase assay (W205GBA2ED: 0.09±0.02, Sulfamylon: 0.08±0.02, Saline: 0.40±0.06, Placebo: 0.45±0.11 μg/mL). *p<0.05, Tukey's multiple comparison test.
  • FIG. 24 shows burn injury upregulates dermal TGF-β expression and treatment with nanoemulsion in the setting of burn wound infection decreases the level of TGF-β in the wound. Dermal levels of the anti-inflammatory cytokines IL-10 and TGF-β were measured in the burn wound 32 hours after thermal injury in animals treated with nanoemulsion, and in animals exposed to bacteria and treated with nanoemulsion or Sulfamylon. A) IL-10 (p=0.4, 1-way ANOVA), and B) TGF-β (p=0.0001). Partial thickness burn increased the presence of dermal TGF-β as compared to sham (624±55 vs. 232±17 pg/mL). Treatment of an infected burn wound with W205GBA2ED significantly reduced the dermal level of TGF-β compared to the untreated burn group (404±43 vs. 624±55 pg/mL). Sulfamylon treatment did not suppress the level of IL-10 or TGF-β in the infected burn wound. *p<0.05, Tukey's multiple comparison test.
  • FIG. 25 shows cross sectional histology of burn skin following infection with P. aeruginosa and treatment with saline or nanoemulsion. Both views of skin are 32 hours after burn injury. A) Representative section from saline (control) treated animal (Hematoxylin and eosin x 40). B) Representative cross-section from W205GBA2ED (nanoemulsion) treated animal (Hematoxylin and eosin x 40).
  • FIG. 26 shows evans blue assay to quantify capillary leak and tissue edema. Evans blue is a dye that binds to serum albumin and can be quantitated to determine vascular permeability. Topical nanoemulsion treatment resulted in less capillary leak following thermal injury and bacterial inoculation of the wound when compared to saline treated control animals (1.26±0.05 vs. 1.93±0.24 μg Evans blue/mg tissue, n=8 per group). *p=0.02, t-test.
  • FIG. 27 shows photomicrographs of partial thickness burned skin with fluorescence labeled TUNEL staining to detect hair follicle cell apoptosis. Skin samples were harvested at 12 hours post-burn. Treatment was performed at 0 and 8 hours following thermal injury. All images are at 40× magnification.
  • FIG. 28 shows TUNEL assay for hair follicle cell apoptosis following a partial thickness burn injury. Skin samples from sham, burn+saline (control), burn+placebo (W205GBA2ED without benzalkonium chloride), and burn+W205GBA2ED were sectioned for fluorescein-labeled TUNEL assay. Treatment was performed at time 0 and 8 hours post injury. No bacterial inoculation of the burned skin was done in this experiment. There was a significant reduction in the degree of hair follicle apoptosis among burn injured and treated animals for tissue samples harvested at 12 hours post injury (p=0.006, 1-way ANOVA). Differences were found for sham vs. burn+saline, burn+saline vs. burn+placebo, and burn+saline vs. burn+W205GBA2ED (*p<0.05, Tukey's multiple comparison test).
  • FIG. 29 shows characterization of the OMP preparation. A) Agarose gel electrophoresis and ethidium bromide-staining. Lane 1. DNA ladder; Lane 2. Whole Cell lysate (WCL) before separation by high speed centrifugation; Lane 3. Supernatant following the 100,000×g spin (lane B in FIGS. 1B, 1C, & 1D); Lane 4. Crude OMP preparation (lane C in FIGS. 1B, 1C, & 1D); Lane 5. Endotoxin (ET) depleted OMP fraction (lane E in FIGS. 1B, 1C, & 1D). Volumetrically-loaded silver stain B) and western blot of OMP preparation (C-D). Lane A. Protein from the supernatant produced after the 6000×g centrifugation was loaded; Lane B. An equal volume of the supernatant from the 100,000×g centrifugation; Lane C. Crude OMP (the re-suspended pellet fraction of the 100,000×g spin); Lanes D-J. Endotoxin-depleted OMP fractions (the successive flow-through portions of the endotoxin column); Lanes K-N. Endotoxin column retentant fractions (the successive fluid regenerated from the column after the addition of sodium deoxycholate). C) Western blot probed with serum from mice immunized with the endotoxin-depleted OMP-NE preparation. D) Western blot probed with serum from mice immunized with the OMP in PBS preparation.
  • FIG. 30 shows antibody responses against B. cenocepacia OMP. A) ELISA results of the IgG response in serum post-immunization with the OMP preparation with or without nanoemulsion. Serum anti-OMP IgG antibody concentrations are presented as mean of endpoint titers in individual sera±SEM. * indicates a statistical difference (p<0.05) in the anti-OMP IgG titers. B) Mucosal antibodies sIgA and IgG against the OMP after nasal vaccine with or without nanoemulsion. sIgA and IgG were measured in BAL solution. The OD levels were normalized to total protein content within the samples.
  • FIG. 31 shows type of cellular immune responses induced by nasal OMP-NE vaccine. A) Serum from mice immunized with 5 μg OMP mixed with either NE (OMP-NE) or with PBS (OMP-PBS) was analyzed for antibody subtype distribution. The results are presented as ratio of the specific subclass IgG to the overall IgG titer. *: indicates statistical difference (p<0.05) between IgG2b and IgG1 subtypes. B) Cytokine profiling of splenocytes of mice immunized with 5 μg. mixed with either NE (OMP-NE) or with PBS (OMP-PBS) Data is represented as fold change±SEM comparing OMP-activated versus non activated splenocytes and is normalized to responses in non-vaccinated mice. *: indicates statistical difference (p<0.05) between OMP-NE and OMP in PBS groups.
  • FIG. 32 shows identification of protein epitopes and LPS in OMP preparations. Lane 1. Protein ladder; Lane 2. Crude OMP preparation; Lane 3. Proteinase K digested crude OMP preparation; Lane 4. Endotoxin-depleted OMP; Lane 5. Proteinase K digested endotoxin-depleted OMP. Western blots were probed with serum from either OMP in PBS or OMP-NE immunized mice or with a monoclonal anti-Pseudomonas mallei LPS antibody as indicated.
  • FIG. 33 shows serum neutralization assay. Percent reduction of B. cenocepacia or B. multivorans cfu plotted against samples with naïve serum. A statistical difference in B. cenocepacia neutralizing activity was observed between serum from mice immunized with OMP-NE and serum from mice immunized with OMP in PBS formulations (p=9.9×10−5 for 5 ug OMP-NE and 0.03 for 15 ug OMP-NE). B. multivorans cross-neutralizing activity was observed between serum from mice immunized with OMP-NE and serum from naïve mice (p=0.04). * indicates a statistically significant (p<0.05) difference in neutralizing activity between OMP-NE and OMP in PBS. ** indicates a statistically significant (p<0.05) difference in neutralizing activity between OMP-NE vaccinated and naïve mice.
  • FIG. 34 shows pulmonary and splenic colonization assay. A) Pulmonary tissue associated and B) Splenic tissue associated cfu determined at six days following intratracheal challenge of 5×107 cfu of B. cenocepacia.
  • DEFINITIONS
  • To facilitate an understanding of the present invention, a number of terms and phrases are defined below:
  • As used herein, the term “microorganism” refers to any species or type of microorganism, including but not limited to, bacteria, viruses, archaea, fungi, protozoans, mycoplasma, prions, and parasitic organisms. The term microorganism encompasses both those organisms that are in and of themselves pathogenic to another organism (e.g., animals, including humans, and plants) and those organisms that produce agents that are pathogenic to another organism, while the organism itself is not directly pathogenic or infective to the other organism.
  • As used herein, the term “pathogen” refers a biological agent that causes a disease state (e.g., infection, sepsis, etc.) in a host. “Pathogens” include, but are not limited to, viruses, bacteria, archaea, fungi, protozoans, mycoplasma, prions, and parasitic organisms.
  • The terms “bacteria” and “bacterium” refer to all prokaryotic organisms, including those within all of the phyla in the Kingdom Procaryotae. It is intended that the term encompass all microorganisms considered to be bacteria including Mycoplasma, Chlamydia, Actinomyces, Streptomyces, and Rickettsia. All forms of bacteria are included within this definition including cocci, bacilli, spirochetes, spheroplasts, protoplasts, etc. Also included within this term are prokaryotic organisms that are Gram-negative or Gram-positive. “Gram-negative” and “Gram-positive” refer to staining patterns with the Gram-staining process, which is well known in the art. (See e.g., Finegold and Martin, Diagnostic Microbiology, 6th Ed., CV Mosby St. Louis, pp. 13-15 (1982)). “Gram-positive bacteria” are bacteria that retain the primary dye used in the Gram stain, causing the stained cells to generally appear dark blue to purple under the microscope. “Gram-negative bacteria” do not retain the primary dye used in the Gram stain, but are stained by the counterstain. Thus, Gram-negative bacteria generally appear red. In some embodiments, bacteria are continuously cultured. In some embodiments, bacteria are uncultured and existing in their natural environment (e.g., at the site of a wound or infection) or obtained from patient tissues (e.g., via a biopsy). Bacteria may exhibit pathological growth or proliferation. Examples of bacteria include, but are not limited to, bacterial cells of a genus of bacteria selected from the group comprising Salmonella, Shigella, Escherichia, Enterobacter, Serratia, Proteus, Yersinia, Citrobacter, Edwardsiella, Providencia, Klebsiella, Hafnia, Ewingella, Kluyvera, Morganella, Planococcus, Stomatococcus, Micrococcus, Staphylococcus, Vibrio, Aeromonas, Plessiomonas, Haemophilus, Actinobacillus, Pasteurella, Mycoplasma, Ureaplasma, Rickettsia, Coxiella, Rochalimaea, Ehrlichia, Streptococcus, Enterococcus, Aerococcus, Gemella, Lactococcus, Leuconostoc, Pedicoccus, Bacillus, Corynebacterium, Arcanobacterium, Actinomyces, Rhodococcus, Listeria, Erysipelothrix, Gardnerella, Neisseria, Campylobacter, Arcobacter, Wolinella, Helicobacter, Achromobacter, Acinetobacter, Agrobacterium, Alcaligenes, Chryseomonas, Comamonas, Eikenella, Flavimonas, Flavobacterium, Moraxella, Oligella, Pseudomonas, Shewanella, Weeksella, Xanthomonas, Bordetella, Franciesella, Brucella, Legionella, Afipia, Bartonella, Calymmatobacterium, Cardiobacterium, Streptobacillus, Spirillum, Peptostreptococcus, Peptococcus, Sarcinia, Coprococcus, Ruminococcus, Propionibacterium, Mobiluncus, Bifidobacterium, Eubacterium, Lactobacillus, Rothia, Clostridium, Bacteroides, Porphyromonas, Prevotella, Fusobacterium, Bilophila, Leptotrichia, Wolinella, Acidaminococcus, Megasphaera, Veilonella, Norcardia, Actinomadura, Norcardiopsis, Streptomyces, Micropolysporas, Thermoactinomycetes, Mycobacterium, Treponema, Borrelia, Leptospira, and Chlamydiae.
  • As used herein, the terms “microorganism” and “microbe” refer to any species or type of microorganism, including but not limited to, bacteria, archaea, fungi, protozoans, mycoplasma, and parasitic organisms.
  • As used herein, the term “fungi” is used in reference to eukaryotic organisms such as molds and yeasts, including dimorphic fungi.
  • As used herein the terms “disease” and “pathologic condition” are used interchangeably, unless indicated otherwise herein, to describe a deviation from the condition regarded as normal or average for members of a species or group (e.g., humans), and which is detrimental to an affected individual under conditions that are not inimical to the majority of individuals of that species or group. Such a deviation can manifest as a state, signs, and/or symptoms (e.g., diarrhea, nausea, fever, pain, blisters, boils, rash, immune suppression, inflammation, etc.) that are associated with any impairment of the normal state of a subject or of any of its organs or tissues that interrupts or modifies the performance of normal functions. A disease or pathological condition may be caused by or result from contact with a microorganism (e.g., a pathogen or other infective agent (e.g., a virus or bacteria)), may be responsive to environmental factors (e.g., malnutrition, industrial hazards, and/or climate), may be responsive to an inherent defect of the organism (e.g., genetic anomalies) or to combinations of these and other factors.
  • “Respiratory” and “respiration” refer to the process by which oxygen is taken into the body and carbon dioxide is discharged, through the bodily system including the nose, throat, larynx, trachea, bronchi and lungs.
  • “Respiratory infection” and “pulmonary infection” refer to an infection (e.g., bacterial, viral, fungal, etc.) of the respiratory tract. In humans, the respiratory tract comprises the upper respiratory tract (e.g., nose, throat or pharynx, and larynx); the airways (e.g.,: voice box or larynx, windpipe or trachea, and bronchi); and the lungs (e.g., bronchi, bronchioles, alveolar ducts, alveolar sacs, and alveoli).
  • “Respiratory disease”, “pulmonary disease,” “respiratory disorder”, “pulmonary disorder,” “respiratory condition”, “pulmonary condition,” “pulmonary syndrome,” and “respiratory syndrome” refer to any one of several ailments that involve inflammation and affect a component of the respiratory system including especially the trachea, bronchi and lungs. Examples of such ailments include acute alveolar disease, obstructive respiratory disease (e.g., asthma; bronchitis; and chronic obstructive pulmonary disease, referred to as COPD), upper airway disease (e.g., such as otitis media, and rhinitis/sinusitis), insterstitial lung disease, allergy, and respiratory infection (e.g., pneumonia, pneyumocystis carinii, and respiratory syncitial virus (RSV)).
  • Specific examples of acute alveolar disease include acute lung injury (ALI), acute respiratory distress syndrome (ARDS), meconium aspiration syndrome (MAS) and respiratory distress syndrome (RDS). ALI is associated with conditions that either directly or indirectly injure the air sacs of the lung, the alveoli. ALI is a syndrome of inflammation and increased permeability of the lungs with an associated breakdown of the lungs' surfactant layer. The most serious manifestation of ALI is ARDS. Among the causes of ALI are complications typically associated with certain major surgeries, mechanical ventilator induced lung injury (often referred to as VILI), smoke inhalation, pneumonia, and sepsis.
  • The term “subject” as used herein refers to organisms to be treated by the compositions of the present invention. Such organisms include animals (domesticated animal species, wild animals), and humans.
  • As used herein, the terms “inactivating,” “inactivation” and grammatical equivalents, when used in reference to a microorganism refer to the killing, elimination, neutralization and/or reducing the capacity of the microorganism to infect and/or cause a pathological response and/or disease in a host.
  • As used herein, the term “fusigenic” is intended to refer to an emulsion that is capable of fusing with the membrane of a microbial agent (e.g., a bacterium or bacterial spore). Specific examples of fusigenic emulsions are described herein.
  • As used herein, the term “lysogenic” refers to an emulsion (e.g., a nanoemulsion) that is capable of disrupting the membrane of a microbial agent (e.g., a virus (e.g., viral envelope) or a bacterium, bacterial spore, or bacterial biofilm). In preferred embodiments of the present invention, the presence of a lysogenic and a fusigenic agent in the same composition produces an enhanced inactivating effect compared to either agent alone. Methods and compositions using this improved antimicrobial composition are described in detail herein.
  • The term “nanoemulsion,” as used herein, includes dispersions or droplets, as well as other lipid structures that can form as a result of hydrophobic forces that drive apolar residues (i.e., long hydrocarbon chains) away from water and drive polar head groups toward water, when a water immiscible oily phase is mixed with an aqueous phase. These other lipid structures include, but are not limited to, unilamellar, paucilamellar, and multilamellar lipid vesicles, micelles, and lamellar phases.
  • As used herein, the terms “contact,” “contacted,” “expose,” and “exposed,” when used in reference to a nanoemulsion and a live microorganism, refer to bringing one or more nanoemulsions into contact with a microorganism (e.g., a pathogen) such that the nanoemulsion kill and/or attenuate growth of the microorganism or pathogenic agent, if present. The present invention is not limited by the amount or type of nanoemulsion used for microorganism killing and/or growth attenuation. A variety of nanoemulsion that find use in the present invention are described herein and elsewhere (e.g., nanoemulsions described in U.S. Pat. Apps. 20020045667 and 20040043041, and U.S. Pat. Nos. 6,015,832, 6,506,803, 6,635,676, and 6,559,189, each of which is incorporated herein by reference in its entirety for all purposes). Ratios and amounts of nanoemulsion are contemplated in the present invention including, but not limited to, those described herein (e.g., in Examples 1-4, the Figures associated therewith and FIG. 17).
  • As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term.
  • The term “surfactant” refers to any molecule having both a polar head group, which energetically prefers solvation by water, and a hydrophobic tail which is not well solvated by water. The term “cationic surfactant” refers to a surfactant with a cationic head group. The term “anionic surfactant” refers to a surfactant with an anionic head group.
  • The terms “Hydrophile-Lipophile Balance Index Number” and “HLB Index Number” refer to an index for correlating the chemical structure of surfactant molecules with their surface activity. The HLB Index Number may be calculated by a variety of empirical formulas as described by Meyers, (Meyers, Surfactant Science and Technology, VCH Publishers Inc., New York, pp. 231-245 [1992]), incorporated herein by reference. As used herein, the HLB Index Number of a surfactant is the HLB Index Number assigned to that surfactant in McCutcheon's Volume 1: Emulsifiers and Detergents North American Edition, 1996 (incorporated herein by reference). The HLB Index Number ranges from 0 to about 70 or more for commercial surfactants. Hydrophilic surfactants with high solubility in water and solubilizing properties are at the high end of the scale, while surfactants with low solubility in water which are good solubilizers of water in oils are at the low end of the scale.
  • As used herein the term “interaction enhancers” refers to compounds that act to enhance the interaction of an emulsion with a microorganism (e.g., with a cell wall of a bacteria (e.g., a Gram negative bacteria) or with a viral envelope. Contemplated interaction enhancers include, but are not limited to, chelating agents (e.g., ethylenediaminetetraacetic acid (EDTA), ethylenebis(oxyethylenenitrilo)tetraacetic acid (EGTA), and the like) and certain biological agents (e.g., bovine serum abulmin (BSA) and the like).
  • The terms “buffer” or “buffering agents” refer to materials which when added to a solution, cause the solution to resist changes in pH.
  • The terms “reducing agent” and “electron donor” refer to a material that donates electrons to a second material to reduce the oxidation state of one or more of the second material's atoms.
  • The term “monovalent salt” refers to any salt in which the metal (e.g., Na, K, or Li) has a net 1+ charge in solution (i.e., one more proton than electron).
  • The term “divalent salt” refers to any salt in which a metal (e.g., Mg, Ca, or Sr) has a net 2+ charge in solution.
  • The terms “chelator” or “chelating agent” refer to any materials having more than one atom with a lone pair of electrons that are available to bond to a metal ion.
  • The term “solution” refers to an aqueous or non-aqueous mixture.
  • As used herein, the term “effective amount” refers to the amount of a composition (e.g., a composition comprising a nanoemulsion) sufficient to effect a beneficial or desired result (e.g., to treat and/or prevent infection (e.g., through bacterial cell killing and/or prevention of bacterial cell growth). An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • As used herein, the term “a composition for inducing an immune response” refers to a composition that, when administered to a subject (e.g., once, twice, three times or more (e.g., separated by weeks, months or years)), stimulates, generates and/or elicits an immune response in the subject (e.g., resulting in total or partial immunity to a microorganism (e.g., pathogen) capable of causing disease). In preferred embodiments of the invention, the composition comprises a nanoemulsion and an immunogen. In further preferred embodiments, the composition comprising a nanoemulsion and an immunogen comprises one or more other compounds or agents including, but not limited to, therapeutic agents, physiologically tolerable liquids, gels, carriers, diluents, adjuvants, excipients, salicylates, steroids, immunosuppressants, immunostimulants, antibodies, cytokines, antibiotics, binders, fillers, preservatives, stabilizing agents, emulsifiers, and/or buffers. An immune response may be an innate (e.g., a non-specific) immune response or a learned (e.g., acquired) immune response (e.g. that decreases the infectivity, morbidity, or onset of mortality in a subject (e.g., caused by exposure to a pathogenic microorganism) or that prevents infectivity, morbidity, or onset of mortality in a subject (e.g., caused by exposure to a pathogenic microorganism)). Thus, in some preferred embodiments, a composition comprising a nanoemulsion and an immunogen is administered to a subject as a vaccine (e.g., to prevent or attenuate a disease (e.g., by providing to the subject total or partial immunity against the disease or the total or partial attenuation (e.g., suppression) of a sign, symptom or condition of the disease.
  • As used herein, the term “adjuvant” refers to any substance that can stimulate an immune response (e.g., a mucosal immune response). Some adjuvants cause activation of a cell of the immune system (e.g., an adjuvant can cause an immune cell to produce and secrete a cytokine). Examples of adjuvants that can cause activation of a cell of the immune system include, but are not limited to, the nanoemulsion formulations described herein, saponins purified from the bark of the Q. saponaria tree, such as QS21 (a glycolipid that elutes in the 21st peak with HPLC fractionation; Aquila Biopharmaceuticals, Inc., Worcester, Mass.); poly(di(carboxylatophenoxy)phosphazene (PCPP polymer; Virus Research Institute, USA); derivatives of lipopolysaccharides such as monophosphoryl lipid A (MPL; Ribi ImmunoChem Research, Inc., Hamilton, Mont.), muramyl dipeptide (MDP; Ribi) and threonyl-muramyl dipeptide (t-MDP; Ribi); OM-174 (a glucosamine disaccharide related to lipid A; OM Pharma SA, Meyrin, Switzerland); cholera toxin (CT), and Leishmania elongation factor (a purified Leishmania protein; Corixa Corporation, Seattle, Wash.). Traditional adjuvants are well known in the art and include, for example, aluminum phosphate or hydroxide salts (“alum”). In some embodiments, immunogenic compositions described herein are administered with one or more adjuvants (e.g., to skew the immune response towards a Th1 and/or Th2 type response).
  • As used herein, the term “an amount effective to induce an immune response” (e.g., of a composition for inducing an immune response), refers to the dosage level required (e.g., when administered to a subject) to stimulate, generate and/or elicit an immune response in the subject. An effective amount can be administered in one or more administrations (e.g., via the same or different route), applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • As used herein, the term “under conditions such that said subject generates an immune response” refers to any qualitative or quantitative induction, generation, and/or stimulation of an immune response (e.g., innate or acquired).
  • As used herein, the term “immune response” refers to any detectable response by the immune system of a subject. For example, immune responses include, but are not limited to, an alteration (e.g., increase) in Toll receptor activation, lymphokine (e.g., cytokine (e.g., Th1 or Th2 type cytokines) or chemokine) expression and/or secretion, macrophage activation, dendritic cell activation, T cell (e.g., CD4+ or CD8+T cell) activation, NK cell activation, and/or B cell activation (e.g., antibody generation and/or secretion). Additional examples of immune responses include binding of an immunogen (e.g., antigen (e.g., immunogenic polypeptide)) to an MHC molecule and induction of a cytotoxic T lymphocyte (“CTL”) response, induction of a B cell response (e.g., antibody production), and/or T-helper lymphocyte response, and/or a delayed type hypersensitivity (DTH) response (e.g., against the antigen from which an immunogenic polypeptide is derived), expansion (e.g., growth of a population of cells) of cells of the immune system (e.g., T cells, B cells (e.g., of any stage of development (e.g., plasma cells), and increased processing and presentation of antigen by antigen presenting cells. An immune response may be to immunogens that the subject's immune system recognizes as foreign (e.g., non-self antigens from microorganisms (e.g., pathogens), or self-antigens recognized as foreign). Thus, it is to be understood that, as used herein, “immune response” refers to any type of immune response, including, but not limited to, innate immune responses (e.g., activation of Toll receptor signaling cascade) cell-mediated immune responses (e.g., responses mediated by T cells (e.g., antigen-specific T cells) and non-specific cells of the immune system) and humoral immune responses (e.g., responses mediated by B cells (e.g., via generation and secretion of antibodies into the plasma, lymph, and/or tissue fluids). The term “immune response” is meant to encompass all aspects of the capability of a subject's immune system to respond to an antigen and/or immunogen (e.g., both the initial response to an immunogen (e.g., a pathogen) as well as acquired (e.g., memory) responses that are a result of an adaptive immune response).
  • As used herein, the term “immunity” refers to protection from disease (e.g., preventing or attenuating (e.g., suppression of) a sign, symptom or condition of the disease) upon exposure to a microorganism (e.g., pathogen) capable of causing the disease. Immunity can be innate (e.g., non-adaptive (e.g., non-acquired) immune responses that exist in the absence of a previous exposure to an antigen) and/or acquired (e.g., immune responses that are mediated by B and T cells following a previous exposure to antigen (e.g., that exhibit increased specificity and reactivity to the antigen)).
  • As used herein, the terms “immunogen” and “antigen” are used interchangeably to refer to an agent (e.g., a microorganism (e.g., bacterium, virus or fungus) and/or portion or component thereof (e.g., a protein antigen)) that is capable of eliciting an immune response in a subject. In preferred embodiments, immunogens elicit immunity against the immunogen (e.g., microorganism (e.g., pathogen or a pathogen product)) when administered in combination with a nanoemulsion of the present invention. As used herein, the term Burkholderia antigen refers to a component or product of a bacteria of the genus Burkholderia that elicits an immune response when administered to a subject. An antigen may be a component or product derived from an organism (e.g., bacteria of the genus Burkholderia) including, but not limited to, polypeptides, peptides, proteins, nucleic acids, membrane fractions, and polysaccharides.
  • As used herein, the term “enhanced immunity” refers to an increase in the level of adaptive and/or acquired immunity in a subject to a given immunogen (e.g., microorganism (e.g., pathogen)) following administration of a composition relative to the level of adaptive and/or acquired immunity in a subject that has not been administered the composition.
  • As used herein, the terms “purified” or “to purify” refer to the removal of contaminants or undesired compounds from a sample or composition. As used herein, the term “substantially purified” refers to the removal of from about 70 to 90%, up to 100%, of the contaminants or undesired compounds from a sample or composition.
  • As used herein, the terms “administration” and “administering” refer to the act of giving a drug, prodrug, or other agent, or therapeutic treatment (e.g., a composition of the present invention) to a physiological system (e.g., a subject or in vivo, in vitro, or ex vivo cells, tissues, and organs).
  • As used herein, the terms “co-administration” and “co-administering” refer to the administration of at least two agent(s) (e.g., a nanoemulsion and one or more other pharmaceutically acceptable substances (e.g., a second nanoemulsion)) or therapies to a subject. In some embodiments, the co-administration of two or more agents or therapies is concurrent. In other embodiments, a first agent/therapy is administered prior to a second agent/therapy. In some embodiments, co-administration can be via the same or different route of administration. Those of skill in the art understand that the formulations and/or routes of administration of the various agents or therapies used may vary. The appropriate dosage for co-administration can be readily determined by one skilled in the art. In some embodiments, when agents or therapies are co-administered, the respective agents or therapies are administered at lower dosages than appropriate for their administration alone. Thus, co-administration is especially desirable in embodiments where the co-administration of the agents or therapies lowers the requisite dosage of a potentially harmful (e.g., toxic) agent(s), and/or when co-administration of two or more agents results in sensitization of a subject to beneficial effects of one of the agents via co-administration of the other agent. In other embodiments, co-administration is preferable to treat and/or prevent infection by more than one type of infectious agent (e.g., bacteria and/or viruses).
  • As used herein, the term “topically” refers to application of a compositions of the present invention (e.g., a composition comprising a nanoemulsion) to the surface of the skin and/or mucosal cells and tissues (e.g., alveolar, buccal, lingual, masticatory, vaginal or nasal mucosa, and other tissues and cells which line hollow organs or body cavities). Compositions described herein can be applied using any pharmaceutically acceptable method, such as for example, intranasal, buccal, sublingual, oral, rectal, ocular, parenteral (intravenously, intradermally, intramuscularly, subcutaneously, intracisternally, intraperitoneally), pulmonary, intravaginal, locally administered, topically administered, mucosally administered, via an aerosol, or via a buccal or nasal spray formulation. Further, the nanoemulsion vaccines described herein can be formulated into any pharmaceutically acceptable dosage form, such as a liquid dispersion, gel, aerosol, pulmonary aerosol, nasal aerosol, ointment, cream, semi-solid dosage form, and a suspension. Further, the composition may be a controlled release formulation, sustained release formulation, immediate release formulation, or any combination thereof.
  • The terms “pharmaceutically acceptable” or “pharmacologically acceptable,” as used herein, refer to compositions that do not substantially produce adverse allergic or immunological reactions when administered to a host (e.g., an animal or a human). Such formulations include dips, sprays, seed dressings, stem injections, sprays, and mists. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, wetting agents (e.g., sodium lauryl sulfate), isotonic and absorption delaying agents, disintegrants (e.g., potato starch or sodium starch glycolate), and the like. Examples of carriers, stabilizers and adjuvants have been described and are known in the art (See e.g., Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, Pa. (1975), incorporated herein by reference).
  • As used herein, the term “pharmaceutically acceptable salt” refers to any salt (e.g., obtained by reaction with an acid or a base) of a composition of the present invention that is physiologically tolerated in the target subject. “Salts” of the compositions of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, sulfonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compositions of the invention and their pharmaceutically acceptable acid addition salts.
  • Examples of bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW4 +, wherein W is C1-4 alkyl, and the like.
  • Examples of salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, chloride, bromide, iodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, undecanoate, and the like. Other examples of salts include anions of the compounds of the present invention compounded with a suitable cation such as Na+, NH4 +, and NW4 + (wherein W is a C1-4 alkyl group), and the like. For therapeutic use, salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
  • For therapeutic use, salts of the compositions of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable composition.
  • As used herein, the terms “at risk for disease” and “at risk for infection” refer to a subject that is predisposed to experiencing a particular disease and/or infection. This predisposition may be genetic (e.g., a particular genetic tendency to experience the disease, such as heritable disorders), or due to other factors (e.g., environmental conditions, exposures to detrimental compounds present in the environment, etc.). Thus, it is not intended that the present invention be limited to any particular risk (e.g., a subject may be “at risk for disease” simply by being exposed to and interacting with other people that carry a risk of transmitting a pathogen), nor is it intended that the present invention be limited to any particular disease and/or infection.
  • “Nasal application”, as used herein, means applied through the nose into the nasal or sinus passages or both. The application may, for example, be done by drops, sprays, mists, coatings or mixtures thereof applied to the nasal and sinus passages.
  • “Pulmonary application” and “pulmonary administration” refers to any means of applying a composition of the present invention to the pulmonary system of a subjet. The present invention is not limited to any particular means of administration. Indeed, a variety of means are contemplated to be useful for pulmonary administration including those described herein.
  • As used herein, the term “kit” refers to any delivery system for delivering materials. In the context of the nanoemulsion compositions of the present invention, such delivery systems include systems that allow for the storage, transport, or delivery of the compositions and/or supporting materials (e.g., written instructions for using the materials, etc.) from one location to another. For example, kits include one or more enclosures (e.g., boxes) containing the relevant nanoemulsions and/or supporting materials. As used herein, the term “fragmented kit” refers to delivery systems comprising two or more separate containers that each contain a subportion of the total kit components. The containers may be delivered to the intended recipient together or separately. For example, a first container may contain a composition comprising a nanoemulsion for a particular use, while a second container contains a second agent (e.g., an antibiotic or spray applicator). Indeed, any delivery system comprising two or more separate containers that each contains a subportion of the total kit components are included in the term “fragmented kit.” In contrast, a “combined kit” refers to a delivery system containing all of the components of a composition needed for a particular use in a single container (e.g., in a single box housing each of the desired components). The term “kit” includes both fragmented and combined kits.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to methods and compositions useful for treating pulmonary infection. In particular, the present invention provides nanoemulsion compositions and methods of using the same to treat bacteria associated with biofilms (e.g., found in pulmonary infections). Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine), industrial, and research applications.
  • Several pathogenic microorganisms initiate infection by attaching to mucosal epithelial cells lining the gastro-intestinal, oropharyngeal, respiratory or genito-urinacy tracts. Some pathogens, such as influenza virus, Bordetella pertussis, or Vibrio cholerae, remain at or within the mucosal tissue, while others, such as Salmonella typhi or hepatitis A virus, possess mechanisms permitting penetration into deeper tissues and spread systemically. Specific and non-specific defense mechanisms of the mucous membranes provide first line protection against both types of pathogen. Non-specific effectors include resident macrophages, antimicrobial peptides, lactoferrin and lysozyme, extremes of pH, bile acids, digestive enzymes, mucus, shedding of epithelial cells, flushing mechanisms (peristalsis, ciliary beating, micturation, etc.) and competition from local flora. However, successful pathogens have generally evolved means to survive the non-specific defenses present at the site they infect and it is the secretory immune system which plays a major role in protecting against diseases caused by a number of bacterial and viral pathogens, and is probably a major effector against pathogens that are restricted to mucosal surfaces. For organisms that spread systemically, both local and systemic immune responses are desirable for optimum immunity.
  • As described herein, certain microbes (e.g., bacteria) are able to thrive when a normal and/or healthy immune system does not function properly for one reason or another. Cystic fibrosis (CF), asthma, HIV infection, chemotherapeutic therapy and a host of other conditions lead to malfunctioning and/or attenuation of immune responses that would normally function to protect against and clear microbes capable of causing pathology in a healthy subject.
  • For example, Pseudomonas aeruginosa is an opportunistic pathogen that infects the immunocompromised, elderly, cancer chemotherapy patients, and individual suffering from CF. In CF lung disease, P. aeruginosa is trapped in thickened, dehydrated, hypoxic mucus lining in airway epithelia. Morphologic data suggests that the airway lumen of CF patients harbor P. aeruginosa biofilms that are characterized as spherical microcolonies.
  • Other types of microbes can cause pathology in an otherwise healthy host subject. For example, colonization of the respiratory tract by the Gram-negative coccobacillus Bordetella pertussis results in whooping cough, also called pertussis, a significant cause of morbidity and mortality of human infants. Two other closely-related isolates of Bordetella have also been found in humans: B. parapertussis and B. bronchiseptica. Molecular genetic analyses suggest that these three isolates are too closely related to be classified as separate species. (See Gilchrist. M. J. R., 1991, “Bordetella”, in Manual of Clinical Microbiology, 5th ed., Balows, A. et al., eds., American Society for Microbiology, Washington, D.C.). While B. pertussis differs from B. bronchiseptica and B. parapertussis in the nature of the toxins it produces, B. bronchiseptica and B. parapertussis do produce active toxins (See Hausman, S. Z. et al., 1996, Infect. Immun. 64: 4020-4026), and there is some evidence to indicate that B. pertussis organisms can covert to the B. parapertussis phenotype (Gilchrist, M. J. R., 1991, “Bordetella”, in Manual of Clinical Microbiology, 5th ed., Balows, A. et al., eds., American Society for Microbiology, Washington, D.C.).
  • Thus, the present invention provides compositions and methods for the treatment of respiratory infections. Compositions and methods of the present invention may be used to treat and/or prevent respiratory infection (e.g., in cystic fibrosis patients) caused by one or more of pseudomonas (e.g., P. aeruginosa, P. paucimobilis, P. putida, P. fluorescens, and P. acidovorans), staphylococci, Methicillin-resistant Staphylococcus aureus (MRSA), streptococci (including Streptococcus pneumoniae), Escherichia coli, Klebsiella, Enterobacter, Serratia, Haemophilus, Yersinia pestis, Burkholderia pseudomallei, B. cepacia, B. gladioli, B. multivorans, B. vietnamiensis, Mycobacterium tuberculosis, M. avium complex (MAC) (M. avium and M. intracellulare), M. kansasii, M. xenopi, M. marinum, M. ulcerans, or M. fortuitum complex (M. fortuitum and M. chelonei), Bordetella pertussis, B. parapertussis and B. bronchiseptica. Furthermore, compositions and methods of the present invention find use in the treatment and/or prevention of a host of respiratory infections (e.g., respiratory infections of the upper respiratory tract (e.g., nose, ears, sinuses, and throat) and the lower respiratory tract (e.g., trachea, bronchial tubes, and lungs)). Several examples of microbes that may be treated (e.g., killed and/or attenuated in growth (e.g., within the respiratory tract of a subject)) are provided below.
  • Accordingly, in some embodiments, the present invention provides a composition comprising a nanoemulsion, and methods of pulmonary administration of the same to prevent and/or treat respiratory infection. In some embodiments, the nanoemulsion comprises ethylenediaminetetraacetic acid (EDTA). The present invention is not limited by the amount of EDTA utilized. In some embodiments, 0.01-0.1 mM, 0.1-1.0 mM, 1.0-1.5 mM, 1.5-2.5 mM, 2.5-5.0 mM, 5.0-10.0 mM, 10-20 mM, 20-30 mM, or 30-50 mM EDTA is used. However, the present invention is not limited to this amount of EDTA. In some embodiments, less than 0.01 mM or more than 50 mM EDTA is utilized. In some embodiments, the composition is co-administered with a hypertonic salt (e.g., sodium chloride) solution (e.g., a 6-7%, 1-3%, 3-6%, 0.1-1%, or more than 7% salt solution (e.g., NaCl solution)). In some embodiments, the composition comprises a 20% nanoemulsion solution. In some embodiments, the composition comprises greater than 20% (e.g., 25%, 30%, or more) nanoemulsion solution. In some embodiments, the composition comprises less than 20% (e.g., 15%, 10% or less) nanoemulsion solution. However, the present invention is not limited to this amount (e.g., percentage) of nanoemulsion. For example, in some embodiments, a composition comprises less than 10% nanoemulsion. In some embodiments, a composition comprises more than 20% nanoemulsion. In some embodiments, the composition comprises 10 mM EDTA. In some embodiments, the composition comprises 20 mM EDTA. In some embodiments, a composition of the present invention comprises any of the nanoemulsions described herein. In some embodiments, a composition comprising a nanoemulsion utilized to treat bacteria (e.g., present in pulmonary space of a subject (e.g., biofilm forming bacteria)) comprises P4075EC. In some embodiments, a composition comprising a nanoemulsion comprises W805EC.
  • Administration of nanoemulsion alone or in combination with EDTA (e.g., 10-20 mM EDTA) was able to achieve complete killing of 106 bacteria in PBS in 60 minutes (See Examples 2-4). In the presence of hypertonic saline (e.g., 6-7% NaCl), the killing ability of the nanoemulsion was surprisingly and strikingly enhanced, achieving complete killing within 15 minutes while in the presence of 20 mM EDTA (See Example 2). Also, nanoemulsions comprising a lower concentration of EDTA were able to achieve complete killing of bacteria in 30 minutes in the presence of hypertonic saline.
  • Thus, in some embodiments, the present invention provides that a nanoemulsion composition can be used to kill (e.g., completely) bacteria over a short time period (e.g., less than 60 minutes, less than 30 minutes, or less than 15 minutes).
  • The present invention also demonstrates that compositions of the present invention are able to eradicate a mixed population of bacteria. Moreover, toxicity studies performed during the development of embodiments of the present invention characterized the nanoemulsion compositions as being safe and causing no detectable harm to a subject (e.g., no histological changes and/or detectable pathology (See, e.g., Example 1)).
  • As described in Examples 3 and 4, compositions comprising nanoemulsions of the present invention are able to treat (e.g., kill and/or inhibit growth of) bacterial species that are generally not virulent in healthy persons, but that are opportunists that cause severe and chronic respiratory tract infections (e.g., in individuals with cystic fibrosis (CF)). Unremitting infection with these species results in inflammation and progressive lung disease that culminates in pulmonary failure, the leading cause of death for CF patients. Effective therapy of pulmonary infection in CF to date has been severely limited by the broad spectrum antimicrobial resistance exhibited by these species, which are among the most drug-resistant bacteria encountered in human infection. The site of infection in CF presents another important obstacle to effective therapy. Infecting bacteria primarily reside within the airway lumen in sputum, airway epithelial surface fluid, and the bronchial mucosa (ref). The penetration of systemically delivered antimicrobials to this infected site is generally poor. Treatment is further hampered by bacterial biofilm formation, which is believed to occur in the airways of infected patients, and by the exceptionally viscous secretions that characterize the CF respiratory tract.
  • Although an understanding of a mechanism is not necessary to practice the present invention, and the present invention is not limited to any particular mechanism of action, in some embodiments, a mechanism of bacterial and/or viral killing utilizing compositions comprising nanoemulsions of the present invention involves fusion of the emulsion with microorganism lipid membranes, leading to rapid osmotic disruption and cell lysis (See, e.g., Hamouda and Baker, J Appl Microbiol. 2000 September; 89(3):397-403). Additionally, in some embodiments, electrostatic attraction (e.g., provided by cationic surface charge of CPC) overcomes the LPS-mediated resistance of gram-negative bacteria to neutral and anionic detergents (See, e.g., Hamouda and Baker, J Appl Microbiol. 2000 September; 89(3):397-403). In some embodiments, bactericidal activity of a composition comprising a nanoemulsion (e.g., P4075EC (e.g., against gram-negative and/or gram-positive bacteria)) is enhanced by the addition of EDTA. Although an understanding of a mechanism is not necessary to practice the present invention, and the present invention is not limited to any particular mechanism of action, in some embodiments, EDTA chelates divalent cations that stabilize outer membrane LPS thereby facilitating interactions with the cationic emulsion, an interaction that leads to membrane permeabilization and lysis as well as augmenting transmembrane diffusion of macromolecules (See, e.g., Rabinovich-Guilatt et al., J Drug Target. 2004; 12(9-10):623-33, Vaara, Microbiol Rev. 1992 September; 56(3):395-411).
  • Experiments conducted during development of embodiments of the present invention identified P4075EC to be stable after nebulization in 7% saline using the PARI LC Plus nebulizer. Inhalation of a single dose of P4075EC nebulized in 7% saline was well tolerated by animals. Thus, in some embodiments, the present invention provides compositions comprising P4075EC and hypertonic saline, and methods of using the same (e.g., via inhalation (e.g., post nebulizing the solution)) for treating one or more types of pulmonary bacterial infections (e.g., to treat bacteria and/or bacterial biofilms (e.g., that possess resistance to conventional treatments) without increasing the already high treatment burden for CF patients).
  • For example, experiments conducted during development of embodiments of the invention show that compositions comprising nanoemulsions (e.g., P4075EC and saline solution) were effective to treat (e.g., kill and/or inhibit growth of) species within the Burkholderia cepacia complex (Bcc) (See, e.g., Example 3). Infection with Burkholderia cepacia species is particularly refractory to antimicrobial therapy and associated with increased rates of morbidity and mortality in CF. Infection with Bcc is also regarded by many CF care centers as an absolute contraindication to lung transplantation. Compositions comprising nanoemulsions (e.g., P4075EC and saline solution) were effective to treat (e.g., kill and/or inhibit growth of) B. multivorans and B. cenocepacia which account for the majority of Bcc infection in CF (See, e.g., Reik J Clin Microbiol. 2005 June; 43(6):2926-8). Compositions comprising nanoemulsions (e.g., P4075EC and saline solution) were effective to treat (e.g., kill and/or inhibit growth of) B. gladioli, which although not a member of the Bcc, is being recovered with increasing frequency from CF patients. Also shown in Example 3, compositions comprising nanoemulsions (e.g., P4075EC) were effective to treat (e.g., kill and/or inhibit growth of) Acinetobacter, which although currently infrequently recovered in CF, appears to be an emerging pathogen in this patient population. The great majority (94%) of isolates tested in Example 3 were recovered from cultures of respiratory specimens from persons with CF. The panel also included one representative isolate from each of five previously described so-called epidemic lineages, each of which has been identified as infecting multiple CF patients. These included the B. cenocepacia ET12 (Johnson et al., J Clin Microbiol 1994; 32:924-30.), PHDC (Chen et al., J Pediatr 2001; 139:643-9), and Midwest (Coenye and LiPuma, J. Infect. Dis. 185:1454-1462) lineages, as well as the B. multivorans OHBM lineage (Biddick et al., FEMS Microbiol Lett 2003; 228:57-62) and the B. dolosa SLC6 lineage (Biddick et al., FEMS Microbiol Lett 2003; 228:57-62). Compositions comprising nanoemulsions (e.g., P4075EC and saline solution) effectively treated (e.g., killed and/or inhibited growth of) each of these different species. Moreover, compositions comprising nanoemulsions (e.g., P4075EC) were effective to treat (e.g., kill and/or inhibit growth of) strains that were found in previous susceptibility testing to be multi-drug resistant (e.g., defined as resistant to all antibiotics tested in two of three antibiotic classes: lactams (including carbapenems), aminoglycosides, and quinolones) as well as panresistant strains (See Example 3). Genotyping analyses of all isolates was performed to confirm that each sample treated was a distinct strain.
  • As shown in Example 3, P4075EC showed very good activity against the strains tested. With the exception of two B. cenocepacia strains, all strains in the test panel were inhibited by a P4075EC concentration of ≦125 μg/ml, or a 1:16 dilution of the starting material; 59% of strains were inhibited by a concentration of ≦31.2 μg/ml, a 1:64 dilution of P4075EC. Multi-drug resistant or panresistant strains did not show decreased susceptibility to P4075EC, demonstrating MIC90s of 125 μg/ml and 62.5 μg/ml, respectively. In general, Burkholderia species tended to be slightly less susceptible to P4075EC than the other species examined. Sixteen of the 18 strains requiring the highest MICs were Burkholderia. Conversely, 33 of the 38 strains with the lowest tested MIC (15.6 μg/ml) were non-Burkholderia species. No striking differences in P4075EC activity was observed among the 10 Burkholderia species examined, although the two strains requiring the highest MICs (250 μg/ml and 500 μg/ml) were both B. cenocepacia. Among the non-Burkholderia species, Ralstonia strains were most susceptible (MIC90≦5.6 μg/ml) while Acinetobacter strains were relatively less susceptible (MIC90=125 μg/ml). No evidence was found of tolerance to P4075EC among a subset of 34 strains for which both MIC and MBC were determined. P4075EC killing of planktonically grown bacteria was time- and concentration-dependent; at a concentration 16 times greater than the MIC, complete killing was achieved within 30 min. Thus, the present invention provides that relatively brief exposure of bacteria to P4075EC can be utilized to effect several log decreases in viable bacteria. Thus, a composition comprising a nanoemulsion (e.g., P4075EC) of the present invention can be utilized individually, or in combination with other antimicrobials, to kill and/or inhibit growth of bacteria.
  • Experiments were conducted during development of embodiments of the invention to assess the activity of P4075EC against bacteria grown in vitro as biofilms. A relatively strict definition of in vitro biofilm formation was employed, as described in Example 3, in an effort to provide a stringent test of P4075EC activity. 12 strains that met the definition were tested. As shown in Example 3, the strains represented several species and a range of susceptibility to P4075EC based on standard MIC/MBC testing. Although the MBIC and MBEC of P4075EC were increased compared to the respective MIC and MBC for each strain tested (median four-fold increase in MBIC compared to MIC), all 12 strains were inhibited or killed by P4075EC when grown as biofilms. Only a single strain required undiluted P4075EC (2000 μg/ml) for eradication of viable biofilm bacteria. Relative biomass was assessed among the biofilm forming strains spectrophotometrically with crystal violet staining and no correlation was observed between biomass and MBIC/MBEC. In fact, although B. gladioli strain AU10529 produced the greatest biomass among the 12 strains tested, the MBIC and MBEC of P4075EC for this strain were relatively low (both 62.5 μg/ml). Conversely, B. cenocepacia strain J2315 produced relatively little biomass, yet required a P4075EC MBEC of 1000 μg/ml.
  • CF sputum, like biofilm, is reported to antagonize the activity of antibacterial drugs (See, e.g., Hunt et al., Antimicrob Agents Chemother. 1995 January; 39(1):34-9). Glycoproteins such as mucin, which compose 2-3% of the dry weight of sputum, and high molecular weight DNA are present in elevated levels resulting in exceptionally viscous sputum that provides a physical barrier protecting bacteria. In addition, these macromolecules bind and sequester antibiotics while small cationic molecules and the decreased pH of CF sputum block drug penetration into bacteria and reduce drug bioactivity. The strategy of increasing drug dosing to overcome these obstacles is limited by drug toxicity. To assess the impact of sputum on the antibacterial activity of P4075EC, standard planktonic susceptibility testing was repeated for the 12 biofilm-forming strains in the presence of CF sputum. A mixture of sputum from 15 CF patients was used to avoid inter-patient variation in macromolecule and high molecular weight DNA composition, and ionic conditions, and only mechanical shearing was applied to minimize changes to the native microenvironment (See, e.g., Grebski et al., Chest. 2001 May; 119(5):1521-5). The activity of P4075EC against bacteria suspended in media containing 43% sputum (the maximum sputum concentration achieved in the test system) was decreased with bactericidal concentrations 2- to 32-fold greater than the respective planktonic MBCs without sputum. The sputum-MBCs were identical to (6 of 12) or within one dilution of (6 of 12) the MBECs obtained with biofilm grown bacteria. Although the activity of P4075EC was similarly antagonized by both CF sputum and biofilm growth, it remained bactericidal for all the strains tested under both test conditions (See FIG. 10).
  • Thus, in some embodiments, the present invention provides compositions comprising nanoemulsions (e.g., P4075EC) and methods of using the same for antimicrobial treatment for infection due to CF-related opportunistic pathogens. In particular, nanoemulsion compositions of the present invention are rapidly bactericidal, and active against bacteria whether grown planktonically or as a biofilm, or in the presence of CF sputum. Moreover, compositions comprising nanoemulsions of the present invention are exceptionally stable, unchanged after nebulization, and broadly microbicidal. Importantly, the development of resistance to a nanoemulsion composition of the present invention has not been observed by any bacterial species examined to date. Thus, the present invention provides that P4075EC can be used effectively as an inhaled antimicrobial.
  • The present invention is not limited to treatment of bacterial biofilms the reside in pulmonary spaces (e.g., within a subject with CF). Indeed, compositions comprising a nanoemulsion of the present invention can be utilized as a therapeutic and/or antimicrobial agent (e.g., to kill and/or inhibit growth of) bacterial biofilms in any clinical and/or industrial setting.
  • Multiple species of bacteria exist that are able to form biofilms. For example, bacteria that adhere to implanted medical devices or damaged tissue often encase themselves in a hydrated matrix of polysaccharide and protein to form biofilm. Biofilms pose a serious problem for public health because of the increased resistance of biofilm-associated organisms to antimicrobial agents and the association of infections with these organisms in patients with indwelling medical devices or damaged tissue. Antibiotic resistance of bacteria growing in biofilms contributes to the persistence and chronic nature of infections such as those associated with implanted medical devices. The mechanisms of resistance in biofilms are different from the now familiar plasmids, transposons, and mutations that confer innate resistance to individual bacterial cells. In biofilms, resistance seems to depend on multicellular strategies.
  • Biofilms are complex communities of microorganisms attached to surfaces or associated with interfaces or damaged tissue. Despite the focus of modern microbiology research on pure culture, planktonic (free-swimming) bacteria, it is now widely recognized that most bacteria found in natural, clinical, and industrial settings persist in association with surfaces as biofilms. Furthermore, these microbial communities are often composed of multiple species that interact with each other and their environment. The determination of biofilm architecture, particularly the spatial arrangement of microcolonies (clusters of cells) relative to one another, has profound implications for the function of these complex communities.
  • The biofilm matrix is a dynamic environment in which the component microbial cells appear to reach homeostasis and are optimally organized to make use of all available nutrients. The matrix therefore shows great microheterogeneity, within which numerous microenvironments can exist. Biofilm formation is believed to be a two-step process in which the attachment of bacterial cells to a surface is followed by growth dependent accumulation of bacteria in multilayered cell clusters. Although exopolysaccharides provide the matrix framework, a wide range of enzyme activities can be found within the biofilm, some of which greatly affect structural integrity and stability.
  • More specifically, during the first phase of formation, it is hypothesized that the fibrinogen and fibronectin of host plasma cover the surface of a medical implant or damaged tissue and are identified by constitutively expressed microbial surface components, which mediate the initial attachment of bacteria to the surface of the biomaterial or damaged tissue. In the second step, a specific gene locus in the bacteria cells, called the intracellular adhesion (ica) locus, activates the adhesion of bacteria cells to each other, forming the secondary layers of the biofilm. The ica locus is responsible for the expression of the capsular polysaccharide operon, which in turn activates polysaccharide intercellular adhesion (PIA), via the sugar poly-N-succinylglucosamine (PNSG), a-1,6-linked glucosaminoglycan. The production of this polysaccharide layer gives the biofilm its slimy appearance when viewed using electron microscopy.
  • Staphylococcus aureus is a highly virulent human pathogen. Both S. aureus and coagulase-negative staphylococci have emerged as major nosocomial pathogens associated with biofilm formation on implanted medical devices and damaged tissue. These organisms are among the normal carriage flora of human skin and mucous membranes, making them prevalent complications during and after invasive surgery or prolonged hospital stays. As bacteria carried on both healthy and sick people, staphylococci are considered opportunistic pathogens that invade patients via open wounds and via biomaterial implants.
  • Biofilm infections associated with S. aureus are a significant cause of morbidity and mortality, particularly in settings such as hospitals, nursing homes and infirmaries. Patients at risk include infants, the elderly, the immuno-compromised, the immuno-suppressed, and those with chronic conditions requiring frequent hospital stays. Patients with intravascular and other implanted prosthetic devices are at even greater risk from staphylococcal infections because of compromised immune systems and the introduction of foreign bodies, which serve to damage tissue and/or act as a surface for the formation of biofilms. Such infections can have chronic, if not fatal, implications.
  • The causes of biofilm resistance to antibiotics include the failure of some antimicrobial agents to penetrate all the layers of a biofilm, the slow-growth rate of certain biofilm cells that make them less susceptible to antimicrobial agents requiring active bacterial growth, and the expression of gene patterns by the bacterial cells embedded in the biofilm that differ from the genes expressed in their planktonic (free-swimming) state. These differences in biofilm-associated bacteria render antimicrobial agents that work effectively to kill planktonic bacteria ineffective in killing biofilm-associated bacteria. Often the only way to treat biofilms (e.g., associated with catheters or prosthetic devices) is the removal of the contaminated device, which may require additional surgery and present further risks to patients.
  • Thus, as used herein, biofilms refer to an aggregate of microorganisms with an extracellular matrix that facilitates adhesion to, and colonization and growth of the aggregate on a surface, such as an internal or external tissue or organ. Biofilms can be comprised of bacteria, fungi, yeast, protozoa, or other microorganisms. Bacterial biofilms typically display high resistance to antibiotics, often up to 1,000-times greater resistance than the same bacteria not growing in a biofilm.
  • In some embodiments, compositions and methods of the invention are utilized to treat (e.g., kill and/or inhibit growth of) and/or prevent biofilms on and/or within a subject (e.g., within the pulmonary system, on internal organs or tissue (e.g., the bladder, kidney, heart, middle ear, sinuses, a joint, the eye), on an external tissue (e.g., the skin), and/or oral surfaces such as teeth, tongue, oral mucosa, or gums. Compositions and methods of the invention may be used to treat a biofilm-associated condition such as a soft-tissue infection, chronic sinusitis, endocarditis, osteomyelitis, urinary tract infection, chronic bacterial vaginosis, dental plaque or halitosis, infection of prosthetic device and/or catheter, bacterial keratitis, or prostatitis.
  • As described in Examples 3 and 4, compositions of the present invention can be utilized to treat (e.g., kill and/or inhibit growth of) any one or more Gram-positive and Gram-negative bacterial species. Indeed, compositions and methods of the present invention can be utilized to kill and/or inhibit growth of a number of bacterial species including, but not limited to, Staphylococcus aureus, coagulase negative staphylococci such as Staphylococcus epidermis, Streptococcus pyogenes (Group A), Streptococcus species (viridans group), Streptococcus agalactiae (group B), S. bovis, Streptococcus (anaerobic species), Streptococcus pneumoniae, Enterococcus species, Bacillus anthracis, Corynebacterium diptheriae, and Corynebacterium species which are diptheroids (aerobic and anaerobic), Listeria monocytogenes, Clostridium tetani, and Clostridium difficile, Escherichia coli, Enterobacter species, Proteus mirablis, Pseudomonas aeruginosa, Klebsiella pneumoniae, Salmonella, Shigella, Serratia, Campylobacter jejuni, Neisseria, Branhamella catarrhalis, and Pasteurella.
  • Compositions and methods of the present invention can be utilized to treat (e.g., kill and/or inhibit growth of) organisms capable of forming biofilms including, but not limited to, dermatophytes (e.g, Microsporum species such as Microsporum canis, Trichophyton species such as Trichophyton rubrum and Trichophyton mentagrophytes), yeasts (e.g., Candida albicans, Candidaparapsilosis, Candida glabrata, Candida tropicalis, and other Candida species including drug resistant Candida species), Epidermophytonfloccosum, Malasseziafuurfur (Pityropsporon orbiculare, Pityropsporon ovale) Cryptococcus neoformans, Aspergillusfumigatus and other Aspergillus species, Zygomycetes (Rizopus, Mucor), hyalohyphomycosis (Fusarium species), Paracoccidiodes brasiliensis, Blastmyces dermatitides, Histoplasma capsulatum, Coccidiodes immitis, Sporothrix schenckii, and Blastomyces.
  • Thus, in some embodiments, the present invention provides a method for treating a subject possessing a biofilm (e.g., possessing an indwelling prosthetic device or catheter, wherein the indwelling prosthetic device or catheter is in contact with a biofilm, or wherein the subject has an infection (e.g., respiratory infection) within which a biofilm resides) comprising administering to the subject a composition comprising a nanoemulsion (e.g., P4075EC) under conditions such that the biofilm is altered and/or bacteria residing within the biofilm are killed and/or their growth is inhibited. In some embodiments, altering the biofilm comprises eradicating the biofilm. In some embodiments, altering the biofilm comprises killing bacteria involved in forming the biofilm. In some embodiments, the bacteria comprise S. aureus, S. epidermidis, antibiotic resistant bacteria (e.g., methicillin resistant, vancomycin resistant, etc.), and/or other type of bacteria described herein. In some embodiments, the composition comprising a nanoemulsion (e.g., P4075EC) is co-administered with one or more antibacterial agents. In some embodiments, the antibacterial agents are selected from the group comprising, but not limited to, antibiotics, antibodies, antibacterial enzymes, peptides, and lanthione-containing molecules. In some embodiments, the antibiotic interferes with or inhibits cell wall synthesis. In some embodiments, the antibiotic is selected from the group including, but not limited to, β-lactams, cephalosporins, glycopeptides, aminoglycosides, sulfonomides, macrolides, folates, polypeptides and combinations thereof. In some embodiments, the antibiotic interferes with protein synthesis (e.g., glycosides, tetracyclines and streptogramins). The present invention is not limited by the number of doses of composition comprising nanoemulsion administered. In some embodiments, multiple doses are administered on separate days. In some embodiments, the multiple doses are administered on the same day. In some embodiments, a composition comprising a nanoemulsion described herein is administered continuously. In some embodiments, co-administration with a composition comprising a nanoemulsion permits administering a lower dose of an antibacterial agent than would be administered without co-administration of a composition comprising a nanoemulsion. In some embodiments, the composition comprising a nanoemulsion described herein is administered using a nebulizer. In some embodiments, administration is intramuscularly, subcutaneously, locally, directly into an infected site, directly onto an indwelling prosthetic device (e.g., a shunt, stent, scaffold for tissue construction, feeding tube, punctual plug, artificial joint, pacemaker, artificial valve, etc.) or catheter. In some embodiments, administration is directly through a catheter.
  • The present invention is not limited by the type of microbe treated. Indeed a variety of microbial pathogens can be treated (e.g, killed (e.g., completely killed)) and/or the growth thereof prevented and/or attenuated in a subject using the compositions and methods of the present invention including, but not limited to, bacteria, viruses, and fungi described herein.
  • The present invention also provides compositions and methods for treating (e.g., killing and/or inhibiting growth of) organisms that heretofore display resistance to a broad spectrum of antibiotics (e.g., species of the genus Acinetobacter).
  • Acinetobacter species are generally considered nonpathogenic to healthy individuals. However, several species persist in hospital environments and cause severe, life-threatening infections in compromised patients (See, e.g., Gerischer U (editor). (2008). Acinetobacter Molecular Biology, 1st ed., Caister Academic Press). The spectrum of antibiotic resistances of these organisms together with their survival capabilities make them a threat to hospitals as documented by recurring outbreaks both in highly developed countries and elsewhere. Infections occur in immunocompromised individuals, and the strain A. baumannii is the second most commonly isolated nonfermenting bacteria in human specimens. Acinetobacter is frequently isolated in nosocomial infections and is especially prevalent in intensive care units, where both sporadic cases as well as epidemic and endemic occurrence is common. A. baumannii is a frequent cause of nosocomial pneumonia, especially of late-onset ventilator associated pneumonia. It can cause various other infections including skin and wound infections, bacteremia, and meningitis. A. lwoffi is also causative of meningitis. A. baumannii can survive on the human skin or dry surfaces for weeks.
  • Since the start of the Iraq War, over 700 U.S. soldiers have been infected or colonized by A. baumannii. Four civilians undergoing treatment for serious illnesses at Walter Reed Army Medical Center in Washington, D.C., contracted A. baumannii infections and died. At Landstuhl Regional Medical Center, a U.S. military hospital in Germany, another civilian under treatment, a 63-year-old German woman, contracted the same strain of A. baumannii infecting troops in the facility and also died.
  • Acinetobacter species are innately resistant to many classes of antibiotics, including penicillin, chloramphenicol, and often aminoglycosides. Resistance to fluoroquinolones has been reported during therapy and this has also resulted in increased resistance to other drug classes mediated through active drug efflux. A dramatic increase in antibiotic resistance in Acinetobacter strains has been reported by the CDC and the carbapenems are recognized as the gold-standard and/or treatment of last resort. An increase in resistance to the carbapenems leaves very little treatment option although there has been some success reported with polymyxin B. Acinetobacter species are unusual in that they are sensitive to sulbactam; sulbactam is most commonly used to inhibit bacterial beta-lactamase, but this is an example of the antibacterial property of sulbactam itself.
  • Thus, in some embodiments, compositions and methods of the present invention are utilized to treat (e.g., kill and/or inhibit growth of) bacteria of the Acinetobacter species (e.g., individually or in combination with other treatments (e.g., carbapenems, polymyxin B, and/or sulbactam)).
  • Cystic Fibrosis
  • Cystic fibrosis (CF) is a life-threatening disorder that causes severe lung damage due to a defective transmembrane protein called CFTR responsible for the balance of electrolytes. Thick mucus forms plugging the tubes, ducts and passageways in the lungs. This environment is ideal for opportunistic bacteria to establish biofilm communities, leading to respiratory infections. Systemically-administered antibiotics can decrease the frequency and severity of exacerbations; however, the bacteria are never be completely eradicated from the airways and the lungs. Nebulized antibiotics are used, but resistance emergence and/or colonization of different resistant species is a major concern. Cystic fibrosis (CF) results in the functional impairment of innate respiratory defense mechanisms, providing an environment for colonization of pathogenic bacterial species such as Staphylococcus aureus and Haemophilus influenzae, and a number of opportunistic species such as Pseudomonas aeruginosa, Achromobacter xylosoxidans, Stenotrophomonas maltophilia, Ralstonia spp., Pandoraea spp., and the Burkholderia cepacia complex (Bcc) species (See, e.g., LiPuma et al., (2009) Antimicrob Agents Chemother 53, 249-255). The Bcc comprises a group of at least 17 phylogenetically related saprophytic gram-negative bacilli, most of which can form biofilm (See, e.g., Al Bakri et al., 2004, Journal of Applied Microbiology 96, 455-463; Eberl and Tummler, 2004, International Journal of Medical Microbiology 294, 123-131; LiPuma et al., (2009) Antimicrob Agents Chemother 53, 249-255; and Tomlin et al., 2004, Journal of Microbiological Methods 57, 95-106). They are particularly difficult to treat and are associated with increased rates of morbidity and mortality in CF patients. They also are among the most antimicrobial-resistant bacterial species encountered in human infections (See, e.g., LiPuma et al, 2005, Curr Opin Pulm Med 11, 528-533; LiPuma et al., (2009) Antimicrob Agents Chemother 53, 249-255). Once established, the infection and associated inflammation are rarely eliminated, resulting in progressive lung disease ending in pulmonary failure and death (See, e.g., LiPuma et al., (2009) Antimicrob Agents Chemother 53, 249-255; Saiman and Seigel, 2003, Infect Control Hosp Epidemiol 24, S6-S52).
  • The present invention is directed to a novel broad-spectrum antimicrobial nanoemulsion (NE) and uses thereof. The NE kills pathogens by interacting with their membranes. This physical kill-on-contact mechanism significantly reduces any concerns about resistance. The NE is formulated from pharmaceutically approved safe ingredients.
  • In Example 8 below, the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of a against four genera of problematic bacteria in CF patients was evaluated: Pseudomonas, Burkholderia, Acinetobacter and Stenotrophomonas. Example 8 also describes evaluating potential synergy between the NE and other antimicrobials. P. aeruginosa, B. cenocepacia, A. baumannii and S. maltophilia, which were evaluated in Example 8, are important respiratory pathogens implicated in acute exacerbations of cystic fibrosis patients. In one aspect of the invention, the activity of a nanoemulsion is defined herein in terms of its minimum inhibitory concentration (MIC), and/or minimum bactericidal concentration (MBC), both in comparison to traditionally used antimicrobial medicines
  • The results described in Example 8 show that the MIC90/MBC90 values for the NE tested were 8/64 μg/ml for P. aeruginosa, 64/>514 μg/ml for B. cenocepacia, 8/64 μg/ml for A. baumannii and 8/32 μg/ml for S. maltophilia. Colistin had MIC90/MBC90 values of 2/8, >32/>32, 1/>16 and >32/>32 for P. aeruginosa, B. cenocepacia, A. baumannii and S. maltophilia, respectively. Cefepime, imipenem, levofloxacin and tobramycin had MIC90/MBC90 values of ≧32/≧32, ≧32/>32, 16/16 and >32/>32 μg/ml, respectively, against all strains. These results are significant as in contrast to conventional drugs such as colistin, cefepime, imipenem, levofloxacin and tobramycin used to treat CF and which can exhibit significant side effects and potential toxicity with increasing dosage, NE are completely non-toxic.
  • Also evaluated in Example 8 below was synergy data. Specifically, also evaluated was the fractional inhibitory concentration (FIC) index when in combination with another antimicrobial and the fractional bactericidal concentration (FBC) index when in combination with another antimicrobial. The FIC and the FBC are calculated to make the judgment on synergy, antagonism or indifference of the nanoemulsion in combination. For this determination, ten strains of Burkholderia, Stenotrophomonas and 10 strains of Acinetobacter were tested to determine a shift in MIC when P4075EC+EDTA was in combination with either colistin or tobramycin, two traditional antimicrobials used in the lungs of CF patients to treat chronic lung infections. The results showed that the NE texted (P4075EC+EDTA) in combination with colistin was found to be synergistic for 90% (in terms of the FIC) and 70% (in terms of the FBC) of the Stenotrophomonas strains, but indifferent, only 20% synergy in by the FIC and 0% by the FBC, when in combination with tobramycin. For the Acinetobacter strains, P4075EC+EDTA in combination with colistin was found to be indifferent, only 20% synergy in by the FIC and 0% by the FBC, as well as when in combination with tobramycin, only 10% synergy in by the FIC and 10% by the FBC. For the Burkholderia strains, P4075EC+EDTA in combination with colistin was found to be indifferent, only 30% synergy in by the FIC and 10% by the FBC, but when in combination with tobramycin, 50% synergy in by the FIC and 20% by the FBC.
  • Finally, Example 8 generated a short time-kill curve to produce samples to be used in pictures using electron microscopy for a strain of Burkholderia to demonstrate the physical kill-on-contact mechanism of action. Time-kill resulted in an overall 4.44 log reduction in cfu/ml from the untreated beginning to the 30 minute time point. Each 10 minute time point had between 1-2 log reduction as follows: from the untreated to the 10 minute point there was a 1.40 log reduction, from the 10 to 20 minute time points there was a 1.91 log reduction and from the 20 to 30 minute time points there was a 1.13 log reduction. See FIGS. 2A-2D.
  • The data herein, such as that presented in Example 8, demonstrates that nanoemulsions, such as the tested P4075EC, are effective against strains that are multidrug-resistant, including colistin-resistant isolates of Burkholderia and Stenotrophomonas. None of the described lipid A modifications in Pseudomonas species impacted the MIC/MBCs with P4075EC. No evidence of antagonism with two major antibiotics, colistin and tobramycin, was observed and in the case of the Stenotrophomonas, synergy was evident. This is valuable because the treatment of patients with CF should not need their normal antibiotic regime suspended in order to use the nanoemulsion, complicating their treatment programs. The SEM images demonstrate the kill on contact mechanism, here in this case a Gram negative bacterium with a reputation of having a tough outer membrane. Further studies are ongoing to investigate the nebulization of P4075EC for the treatment of pulmonary infection in cystic fibrosis patients.
  • While therapeutic progress has been made in preserving functional pulmonary physiology in CF patients by managing nutrition and mucosal secretions, little progress has been made in therapeutic interventions for the prevention and management of Bcc infections. For example, at present, no effective vaccines against Bcc are available. Mucosal vaccine development for Bcc has been limited in part due to the lack of an identified protective antigen and the lack of effective mucosal adjuvants (See, e.g., Davis, 2001, Advanced Drug Delivery Reviews 51, 21-42).
  • As shown in Example 10 below, the present invention provides immunogenic compositions comprising a nanoemulsion and Burkholderia antigen (e.g., Burkholderia outer membrane protein (OMP)) that, when administered to a subject, induces immunity (e.g., protective immunity) in the subject against bacteria from the genus Burkholderia (e.g., B. cenocepacia, B. multivorans or others species associated with respiratory infection). In some embodiments, the present invention provides all or a portion of an OMP (e.g., isolated, purified, and/or recombinant OMPs)) from a bacteria of the genus Burkholderia (See, e.g., Example 10, Table 25) that is utilized in an immunogenic composition comprising a nanoemulsion (e.g., for administration to a subject (e.g., to induce immunity to a bacteria of the genus Burkholderia in the subject)). As shown in Example 10, the present invention provides that when administered to a subject, an immunogenic composition comprising a nanoemulsion and Burkholderia OMP induces both mucosal and systemic anti-OMP antibodies. Also shown in Example 10, the present invention provides that when administered to a subject, an immunogenic composition comprising a nanoemulsion and an OMP from a specific strain of Burkholderia (e.g., B. cenocepacia) induces both mucosal and systemic anti-OMP antibodies that neutralize bacteria of the specific species from which the OMP is derived (e.g., B. cenocepacia) and cross-neutralize bacteria from one or more other species of bacteria (e.g., of the genus Burkholderia).
  • Example 10 shows that a single nasal immunization with OMP-NE produces a robust immune response characterized by rapid induction of antigen-specific T cell responses and secretory antibody responses, as documented by induction of IFN-γ, IL-2 and mucosal sIgA and IgG, and that the immune response can be subsequently boosted. Mice immunized with OMP-NE displayed a dramatic decrease in K56-1 colony forming units (cfu) in the lungs when compared with non-immunized controls. Pulmonary bacterial clearance was significantly enhanced in OMP-NE vaccinated mice which showed fewer B. cenocepacia organisms in the spleens as compared to non-vaccinated groups. Thus, the present invention provides that mucosal OMP-NE administration produced protective immunity and reduced the likelihood of sepsis in subjects.
  • Example 10 also provides data identifying a 17 KDa OmpA-like protein and immune responses associated with exposure to the protein. The 17 KDa lipoprotein fractions were better recognized by antibodies contained in serum from OMP-NE vaccinated mice (See, e.g., Example 10, FIG. 29C). Thus, the present invention provides that immunogenic compositions comprising epitopes contained within the 17 KDa OmpA-like proteins generate protective antibodies (e.g., against one or more species of Burkholderia) in a subject administered the immunogenic composition.
  • Significant differences between OMP-NE and OMP in PBS responses were observed. For example, immunization with OMP in the absence of nanoemulsion resulted in relatively high titers of IgG, but were consistently lower (13 to 30 fold) than those induced by OMP-NE immunogenic compositions. Immunization with OMP in the absence of nanoemulsion failed to respond to a booster immunization. The response induced with OMP plus nanoemulsion was primarily directed toward specific proteins contained within the preparation, whereas OMP in the absence of nanoemulsion appears to have elicited immune responses to the intrinsic endotoxin content.
  • Accordingly, the present invention provides methods and compositions for the stimulation of immune responses. In particular, the present invention provides immunogenic nanoemulsion compositions and methods of using the same for the induction of immune responses (e.g., innate and/or adaptive immune responses (e.g., for generation of host immunity against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans, etc.))). Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine (e.g., vaccination)) and research applications.
  • In some embodiments, the present invention provides nanoemulsion adjuvants and compositions comprising the same (e.g., vaccines) for the stimulation of immune responses (e.g., immunity) against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans, etc.). In some embodiments, the present invention provides nanoemulsion adjuvant compositions that stimulate and/or elicit immune responses (e.g., innate immune responses and/or adaptive/acquired immune responses) when administered to a subject (e.g., a human or other mammalian subject)). In some embodiments, the present invention provides nanoemulsion adjuvant compositions comprising one or a plurality of Burkholderia (e.g., B. cenocepacia, B. multivorans, etc.) antigens (e.g., Burkholderia components isolated and/or purified, and/or recombinant Burkholderia proteins (e.g., OMPs)). The present invention is not limited to any particular nanoemulsion or Burkholderia antigen. Exemplary immunogenic compositions (e.g., vaccine compositions) and methods of administering the compositions are described in more detail below.
  • In some embodiments, the present invention provides an immunogenic composition comprising a nanoemulsion and one or more Burkholderia antigens (e.g., B. cepacia antigens). In some embodiments, the present invention provides a method of inducing an immune response to Burkholderia (e.g., B. cepacia) in a subject comprising: providing a subject and an immunogenic composition comprising a nanoemulsion and an immunogen, wherein the immunogen comprises a Burkholderia (e.g., Burkholderia cepacia) antigen and administering the composition to the subject under conditions such that the subject generates a Burkholderia (e.g., Burkholderia cepacia) specific immune response. The present invention is not limited by the route chosen for administration of a composition of the present invention. In some preferred embodiments, administering the immunogenic composition comprises contacting a mucosal surface of the subject with the composition. In some embodiments, the mucosal surface comprises nasal mucosa. In some embodiments, inducing an immune response induces immunity to Burkholderia (e.g., Burkholderia cepacia) in the subject.
  • Experiments were conducted during development of embodiments of the invention to determine if a composition comprising a nanoemulsion (NE) and Burkholderia antigen could be utilized to generate an immune response in a subject. Nasal immunization with a whole cell Streptococcus pneumoniae antigen (WCPAg) mixed with nanoemulsion was performed and shown to induce an IgG response in a host subject and the ability to eradicate upper respiratory colonization of S. pneumoniae.
  • In particular, as described in Example 10, the present invention provides immunogenic compositions comprising a nanoemulsion and Burkholderia antigen (e.g., Burkholderia outer membrane protein (OMP)) that, when administered to a subject, induces immunity (e.g., protective immunity) in the subject against bacteria from the genus Burkholderia (e.g., B. cenocepacia, B. multivorans or others species associated with respiratory infection). Accordingly, in some embodiments, the present invention provides that administration (e.g., nasal administration) of a composition comprising nanoemulsion and Burkholderia antigen (e.g., OMP antigen (e.g., a 17 KDa protein comprising an amino acid sequence selected from SEQ ID NOs. 1-16)) to a subject produces immunity toward Burkholderia in the subject thereby protecting the subject against Burkholderia infection (e.g., associated with a respiratory infection).
  • The present invention is not limited by the type of bacteria of the genus Burkholderia utilized in the immunogenic compositions and methods of using the same of the invention. In some embodiments, the bacteria is a pathogen. In some embodiments, the pathogen is a Burkholderia species responsible for respiratory or respiratory associated infection. A variety of Burkholderia species find use in the compositions and methods of the invention including, but not limited to, B. cenocepacia, B. dolosa, B. multivorans, B. ambifaria, B. vietnamiensis, B. ubonensis, B. thailandensis, B. graminis, B. oklahomensis, B. pseudomallei, B. xenovorans, B. phytofirmans, B. phymatum, R. metallidurans, R. eutropha, R. solanacearum.
  • In some embodiments, the bacteria of the genus Burkholderia is B. cepecia. In some embodiments, an immunogenic composition comprising a nanoemulsion and an Omp-A like protein comprises an Omp-A like protein from a Burkholderia species including, but not limited to, B. cenocepacia, B. dolosa, B. multivorans, B. ambifaria, B. vietnamiensis, B. ubonensis, B. thailandensis, B. graminis, B. oklahomensis, B. pseudomallei, B. xenovorans, B. phytofirmans, B. phymatum, R. metallidurans, R. eutropha, R. solanacearum. In some embodiments, an immunogenic composition comprising a nanoemulsion and an Omp-A like protein comprises an Omp-A like protein (e.g., isolated, purified, and/or recombinant Omp-A like protein) comprising an amino acid sequence identified in SEQ ID NOs.: 1-16. In some embodiments, an immunogenic composition comprising a nanoemulsion and an Omp-A like protein comprises an Omp-A like protein (e.g., isolated, purified, and/or recombinant Omp-A like protein) comprising an amino acid sequence of SEQ ID NO. 1.
  • In some embodiments, an immunogenic composition comprising a nanoemulsion and Burkholderia antigen comprises antigens (e.g., polysaccharide, protein, killed whole cells (e.g., conjugated or non-conjugated antigens)), wherein the antigens are derived from multiple (e.g., at least 2, 3, 5, 7, 10, 15, 20 or more) serotypes of Burkholderia. The number of Burkholderia antigens utilized can range from 2 different serotypes to about 20 different serotypes. In some embodiments, an immunogenic composition comprising a nanoemulsion and Burkholderia antigen may comprise Burkholderia antigen (e.g., whole cell, polysaccharide, Omp-A protein, other protein, etc.) from every known and/or isolated Burkholderia serotype.
  • In some embodiments, an immunogenic composition comprising a nanoemulsion and Burkholderia (e.g., B. cepacia) antigen comprises one, two or more different types of carrier protein (e.g., that act as carriers for proteins, saccharides, etc.). For example, in one embodiment, two or more different saccharides or proteins may be conjugated to the same carrier protein, either to the same molecule of carrier protein or to different molecules of the same carrier protein. Carrier proteins may be TT, DT, CRM197, fragment C of TT, PhtD, PhtBE or PhtDE fusions (particularly those described in WO 01/98334 and WO 03/54007), detoxified pneumolysin and protein D. In some embodiments, a carrier protein present in a composition comprising a nanoemulsion and Burkholderia (e.g., B. cenocepacia) antigen is a member of the polyhistidine triad family (Pht) proteins, fragments or fusion proteins thereof. The PhtA, PhtB, PhtD or PhtE proteins may have an amino acid sequence sharing 80%, 85%, 90%, 95%, 98%, 99% or 100% identity with a sequence disclosed in WO 00/37105 or WO 00/39299 (e.g. with amino acid sequence 1-838 or 21-838 of SEQ ID NO: 4 of WO 00/37105 for PhtD). For example, fusion proteins are composed of full length or fragments of 2, 3 or 4 of PhtA, PhtB, PhtD, PhtE. Examples of fusion proteins are PhtA/B, PhtA/D, PhtA/E, PhtB/A, PhtB/D, PhtB/E. PhtD/A. PhtD/B, PhtD/E, PhtE/A, PhtE/B and PhtE/D, wherein the proteins are linked with the first mentioned at the N-terminus (see for example WO01/98334). Carriers may comprise histidine triad motif(s) and/or coiled coil regions. A histidine triad motif is the portion of polypeptide that has the sequence HxxHxH where H is histidine and x is an amino acid other than histidine. A coiled coil region is a region predicted by “Coils” algorithm Lupus, A et al (1991) Science 252; 1162-1164.
  • Examples of carrier proteins which may be used in the present invention are DT (Diphtheria toxoid), TT (tetanus toxoid) or fragment C of TT, DT CRM197 (a DT mutant) other DT point mutants, such as CRM176, CRM228, CRM 45 (Uchida et al J. Biol. Chem. 218; 3838-3844, 1973); CRM 9, CRM 45, CRM102, CRM 103 and CRM107 and other mutations described by Nicholls and Youle in Genetically Engineered Toxins, Ed: Frankel, Maecel Dekker Inc, 1992; deletion or mutation of Glu-148 to Asp, Gln or Ser and/or Ala 158 to Gly and other mutations disclosed in U.S. Pat. No. 4,709,017 or U.S. Pat. No. 4,950,740; mutation of at least one or more residues Lys 516, Lys 526, Phe 530 and/or Lys 534 and other mutations disclosed in U.S. Pat. No. 5,917,017 or U.S. Pat. No. 6,455,673; or fragment disclosed in U.S. Pat. No. 5,843,711, pneumococcal pneumolysin (Kuo et al (1995) Infect Immun 63; 2706-13) including ply detoxified in some fashion for example dPLY-GMBS (WO 04081515, PCT/EP2005/010258) or dPLY-formol, PhtX, including PhtA, PhtB, PhtD, PhtE and fusions of Pht proteins for example PhtDE fusions, PhtBE fusions (WO 01/98334 and WO 03/54007), (Pht A-E are described in more detail below) OMPC (meningococcal outer membrane protein—usually extracted from N. meningitidis serogroup B—EP0372501), PorB (from N. meningitidis), PD (Haemophilus influenzae protein D—see, e.g., EP 0 594 610 B), or immunologically functional equivalents thereof, synthetic peptides (EP0378881, EP0427347), heat shock proteins (WO 93/17712, WO 94/03208), pertussis proteins (WO 98/58668, EP0471177), cytokines, lymphokines, growth factors or hormones (WO 91/01146), artificial proteins comprising multiple human CD4+ T cell epitopes from various pathogen derived antigens (Falugi et al (2001) Eur J Immunol 31; 3816-3824) such as N19 protein (Baraldoi et al (2004) Infect Immun 72; 4884-7) pneumococcal surface protein PspA (WO 02/091998), iron uptake proteins (WO 01/72337), toxin A or B of C. difficile (WO 00/61761).
  • Generation of Antibodies
  • An immunogenic composition comprising a nanoemulsion and Burkholderia (e.g., B. cenocepacia) antigen can be used to immunize a mammal, such as a mouse, rat, rabbit, guinea pig, monkey, or human, to produce polyclonal antibodies. If desired, a Burkholderia (e.g., B. cenocepacia) antigen can be conjugated to a carrier protein, such as bovine serum albumin, thyroglobulin, keyhole limpet hemocyanin or other carrier described herein. Depending on the host species, various adjuvants can be used to increase the immunological response. Such adjuvants include, but are not limited to, Freund's adjuvant, mineral gels (e.g., aluminum hydroxide), and surface active substances (e.g. lysolecithin, pluronic polyols, polyanions, peptides, nanoemulsions described herein, keyhole limpet hemocyanin, and dinitrophenol). Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are especially useful.
  • Monoclonal antibodies that specifically bind to a Burkholderia (e.g., B. cepacia) antigen can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These techniques include, but are not limited to, the hybridoma technique, the human B cell hybridoma technique, and the EBV hybridoma technique (See, e.g., Kohler et al., Nature 256, 495 497, 1985; Kozbor et al., J. Immunol. Methods 81, 3142, 1985; Cote et al., Proc. Natl. Acad. Sci. 80, 2026 2030, 1983; Cole et al., Mol. Cell. Biol. 62, 109 120, 1984).
  • In addition, techniques developed for the production of “chimeric antibodies,” the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used (See, e.g., Morrison et al., Proc. Natl. Acad. Sci. 81, 68516855, 1984; Neuberger et al., Nature 312, 604 608, 1984; Takeda et al., Nature 314, 452 454, 1985). Monoclonal and other antibodies also can be “humanized” to prevent a patient from mounting an immune response against the antibody when it is used therapeutically. Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between rodent antibodies and human sequences can be minimized by replacing residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grating of entire complementarity determining regions.
  • Alternatively, humanized antibodies can be produced using recombinant methods, as described below. Antibodies which specifically bind to a particular antigen can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. Pat. No. 5,565,332.
  • Alternatively, techniques described for the production of single chain antibodies can be adapted using methods known in the art to produce single chain antibodies which specifically bind to a particular antigen. Antibodies with related specificity, but of distinct idiotypic composition, can be generated by chain shuffling from random combinatorial immunoglobin libraries (See, e.g., Burton, Proc. Natl. Acad. Sci. 88, 11120 23, 1991).
  • Single-chain antibodies also can be constructed using a DNA amplification method, such as PCR, using hybridoma cDNA as a template (See, e.g., Thirion et al., 1996, Eur. J. Cancer Prey. 5, 507-11). Single-chain antibodies can be mono- or bispecific, and can be bivalent or tetravalent. Construction of tetravalent, bispecific single-chain antibodies is taught, for example, in Coloma & Morrison, 1997, Nat. Biotechnol. 15, 159-63. Construction of bivalent, bispecific single-chain antibodies is taught, for example, in Mallender & Voss, 1994, J. Biol. Chem. 269, 199-206.
  • A nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below. Alternatively, single-chain antibodies can be produced directly using, for example, filamentous phage technology (See, e.g., Verhaar et al., 1995, Int. J. Cancer 61, 497-501; Nicholls et al., 1993, J. Immunol. Meth. 165, 81-91).
  • Antibodies which specifically bind to a particular antigen also can be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature (See, e.g., Orlandi et al., Proc. Natl. Acad. Sci. 86, 3833 3837, 1989; Winter et al., Nature 349, 293 299, 1991).
  • Chimeric antibodies can be constructed as disclosed in WO 93/03151. Binding proteins which are derived from immunoglobulins and which are multivalent and multispecific, such as the “diabodies” described in WO 94/13804, also can be prepared. Antibodies can be purified by methods well known in the art. For example, antibodies can be affinity purified by passage over a column to which the relevant antigen is bound. The bound antibodies can then be eluted from the column using a buffer with a high salt concentration.
  • Burn Wound Management
  • Contemporary burn wound management involves early debridement and reconstruction of non-viable skin coupled with provision of supportive care and topical antimicrobial dressing changes to partial thickness burn wounds. The goal of modern burn wound care is to provide an optimal environment for epidermal renewal. Restoration of skin integrity takes place via regrowth of keratinocytes from preserved hair follicles or transfer of split thickness skin grafts harvested from non-burn regions. During the period of epidermal renewal it is important to avoid further injury to the skin, abrogate burn wound progression, and minimize secondary complications such as wound infection. Early excision of full-thickness burn eschar, immediate skin grafting, and treatment of remaining open or partial thickness areas of burn wound with topical antimicrobial agents is the most effective way of minimizing burn wound colonization and invasive wound infection. (See, e.g., Bessey, Wound care. In Herndon D N, ed: Total Burn Care 3rd edition. Philadelphia, Pa.: Elsevier Inc., 2007, pp 127-135.). Popular topical antimicrobial agents include silver sulfadiazine (SILVADENE), mafenide acetate (SULFAMYLON), and colloidal silver impregnated dressings (ACTICOAT, SILVERLON). Each of these agents has potential limitations such as variable ability to penetrate eschar, uneven efficacy against both Gram-negative and Gram-positive bacteria, and potential toxicity to host immune cells (See, e.g., Steinstraesser et al., Antimicrob Agents Chemother 46(6):1837-1844, 2002).
  • Accordingly, the present invention provides nanoemulsion compositions and methods of using the same for the treatment of burn wounds. For example, as shown in Example 9, the present invention provides nanoemulsion compositions and methods of using the same to reduce, attenuate and/or prevent bacterial growth in a burn wound. The present invention also provides nanoemulsion compositions that reduce wound inflammation following burn injury. Although an understanding of a mechanism of action is not needed to practice the present invention, and the present invention is not limited to any particular mechanism of action, in some embodiments, a nanoemulsion composition that is applied to a wound following burn injury is able to penetrate more deeply and uniformly into a burn wound (e.g., thereby decreasing and/or inhibiting bacterial growth and/or inflammation at the site of the wound). As shown in Example 9, the present invention provides a method of reducing, inhibiting and/or eliminating bacterial growth in a burn wound comprising providing a burn wound and a nanoemulsion and administering the nanoemulsion to the burn wound under conditions that bacterial growth is reduced, inhibited and/or eliminated. In some embodiments, a nanoemulsion composition described herein is combined with one or more antimicrobial drugs for administration to a burn wound to minimize bacterial growth at the burn wound site. The present invention is not limited to any particular antimicrobial drug. Indeed, any antimicrobial drug that inhibits bacterial growth known to those in the art can be utilized in combination with a nanoemulsion composition described herein.
  • In addition to local effects, severe dermal burns are known to induce the systemic inflammatory response syndrome (SIRS), which results in a high-risk of end-organ dysfunction (See, e.g., Barton et al., J Burn Care Rehabil 18(1):1-9, 1997). Increased vascular permeability and systemic capillary leak as a consequence of SIRS following burn injury creates seepage of plasma into interstitial tissue throughout the body. This tissue edema and intravascular hypovolemia is responsible for a host of undesired clinical problems such as shock, pulmonary dysfunction, abdominal or extremity compartment syndrome, and cardiac failure.
  • As shown in Example 9 below, nanoemulsion compositions described herein can be administered to a burn wound to treat (e.g., reduce, attenuate and/or prevent) inflammation, tissue edema and/or intravascular hypovolemia at the site of a burn wound. In some embodiments, reducing inflammation, tissue edema and/or intravascular hypovolemia at the site of a burn wound reduces the occurrence of shock, pulmonary dysfunction, abdominal or extremity compartment syndrome, and/or cardiac failure. In some embodiments, a nanoemulsion composition described herein is used in combination with (e.g., is co-administered with) one or more anti-inflammatory drugs to minimize early burn wound inflammation and tissue edema. The present invention is not limited to any particular anti-inflammatory drug. Indeed, any anti-inflammatory drug that minimizes early burn wound inflammation and tissue edema can be utilized in combination with a nanoemulsion composition described herein.
  • In some embodiments, a nanoemulsion of the invention (e.g., W205GBA2ED) is administered to a burn wound to prevent, attenuate and/or eradicate bacterial growth (e.g., Staphylococcus aureus, P. aeruginosa, or other bacteria) within a partial thickness burn wound. Example 9 shows that reduction in microbial infection was coupled with generation of lower levels of local dermal pro-inflammatory cytokines and evidence of reduced neutrophil sequestration into the burn wound. This decrease in burn wound bacterial growth and inflammation also produced less capillary leak in the early post-thermal injury time-period. Having the ability to clinically reduce capillary leak and tissue edema in the immediate post-burn time-period provides a lesser need for large volume crystalloid fluid resuscitation and a reduction in the associated sequela of physiologic volume overload, pulmonary dysfunction, and abdominal compartment syndrome.
  • Skin that is damaged by thermal injury loses its ability to protect the host against infection from both the loss of physical barrier function and the secondary immunosuppression caused by the thermal injury. Moreover, increased production of TGF-β and IL-10 during the post-burn period can result in immunosuppression. (See, e.g., Lyons et al., Arch Surg 134(12):1317-1323, 1999; Varedi et al., Shock 16(5):380-382, 2001). It has been established that treatment of burn injured animals with anti-TGF-β can improve local and systemic clearance of P. aeruginosa (See, e.g., Huang et al., J Burn Care Res 27(5):682-687, 2006). Inhibition of TGF-β also results in increased survival following bacterial challenge. As shown in Example 9, a significant elevation of TGF-(3, but not IL-10 was observed in the skin following partial thickness burn injury. However, topical nanoemulsion application (e.g., 10% W205GBA2ED) to the burn wound inoculated with bacteria resulted in a reduction of the level of TGF-β when compared to the untreated burn wound.
  • Onset of a bacterial infection within a burn wound can delay or even reverse the tissue healing process (See, e.g., Steinstraesser et al., Crit. Care Med 29(7):1431-1437, 2001). Topical antimicrobial therapy is used to reduce the microbial load in the burn wound and reduce this risk of infection. Current topical agents include silver nitrate (AgNO3), silver sulfadiazine, mafenide acetate, and nanocrystalline impregnated silver dressings. Silver nitrate is limited in its use because of the problem it creates from contact staining and its limited antifungal activity. Silver sulfadiazine is the mainstay of topical burn antimicrobial treatment. It is bactericidal against P. aeruginosa and other Gram-negative enteric bacteria. Resistance to Silvadene by some of these organisms has emerged (See, e.g., Silver et al., J Ind Microbiol Biotechnol 33(7):627-634, 2006). The agent has limited antifungal activity, but can be used in conjunction with nystatin. Silvadene has no real ability to penetrate burn eschar and sometimes leads to leukopenia which requires conversion to another topical agent. The use of mafenide acetate is narrowed by the fact that it is bacteriostatic against select organisms, it has limited activity against Gram-positive bacteria such as Staphylococcus aureus, and that its use over a large surface area can lead to a metabolic acidosis because of its metabolism into a carbonic anhydrase inhibitor. The nanocrystalline silver dressings have the broadest activity against burn wound pathogens of the current agents available. They have a modest ability to penetrate eschar and can be left in place for many days (See, e.g., Church et al., Clin Microbiol Rev 19(2):403-434, 2006). SB 202190, an inhibitor of activated p38 MAPK, can substantially reduce the dermal inflammation generated in burn wounds (See, e.g., Arbabi et al., Shock. 26(2):201-209, 2006). Thermal injury initiates dermal inflammatory and pro-apoptotic cell signaling.
  • As shown in Example 9, topical application of nanoemulsion (e.g., W205GBA2ED resulted in reduced hair follicle cell apoptosis within the dermis of burned skin. Thus, in some embodiments, the present invention provides nanoemulsion compositions that can be utilized to reduce, when administered to a burn wound, conversion of the partial thickness burn wound within the “zone of stasis” to regions of full thickness burn.
  • In patients without evidence of inhalational injury, the burn wound itself is the primary source triggering the systemic inflammatory response via generation of pro-inflammatory cytokines and sequestration of neutrophils into the burn wound (See, e.g., Hansbrough et al., J Surg Res 61(1):17-22, 1996; Piccolo et al., Inflammation 23(4):371-385, 1999; Till et al., J Clin Invest 69(5):1126-1135, 1982). Topical application of a p38 MAPK inhibitor can control the source of inflammation at the level of the dermis, resulting in lower levels of pro-inflammatory mediators, reduced neutrophil sequestration and microvascular damage, and less epithelial apoptosis in burn wound hair follicle cells (See, e.g., Ipaktchi et al., Shock. 26(2):201-209, 2006). Dermal source control of inflammation also reduces bacterial growth and attenuates the systemic inflammatory response resulting in less acute lung injury and cardiac dysfunction following partial thickness burn injury in a rodent model. Accordingly, in some embodiments, a nanoemulsion of the invention is utilized (e.g., administered) alone or in combination with an anti-inflammatory and/or antimicrobial agent to reduce local dermal inflammation and risk of infection within burn wounds (e.g., early burn wounds, partial thickness wounds, full thickness wounds or other burn wounds). The present invention is not limited by the type of anti-inflammatory agent and/or antimicrobial utilized for co-administration with a nanoemulsion described herein. Indeed, a variety of anti-inflammatory agents and/or antimicrobial agents can be used including, but not limited to, silver nitrate (AgNO3), silver sulfadiazine, mafenide acetate, nanocrystalline impregnated silver dressings, p38 MAPK inhibitor (e.g., SB 202190), or another anti-inflammatory or antimicrobial agent described herein.
  • In some embodiments, when a nanoemulsion of the invention is administered to a burn wound, the nanoemulsion can be administered (e.g., to a subject (e.g., to a burn or wound surface)) by multiple methods, including, but not limited to, direct use or being suspended in a solution (e.g., colloidal solution) and applied to a surface (e.g., a surface comprising bacteria (e.g., pathogenic bacteria) or susceptible to bacterial invasion); being sprayed onto a surface using a spray applicator; being mixed with fibrin glue and applied (e.g., sprayed) onto a surface (e.g., skin burn or wound); being impregnated onto a wound dressing or bandage and applying the bandage to a surface (e.g., an infection or burn wound); being applied by a controlled-release mechanism; or being impregnated on one or both sides of an acellular biological matrix that is then placed on a surface (e.g., skin burn or wound) thereby protecting at both the wound and graft interfaces. In some embodiments, the invention provides a pharmaceutical composition containing (a) a composition comprising a nanoemulsion (e.g., W205GBA2ED); and (b) one or more other agents (e.g., an antibiotic). Examples of other types of antibiotics include, but are not limited to, almecillin, amdinocillin, amikacin, amoxicillin, amphomycin, amphotericin B, ampicillin, azacitidine, azaserine, azithromycin, azlocillin, aztreonam, bacampicillin, bacitracin, benzyl penicilloyl-polylysine, bleomycin, candicidin, capreomycin, carbenicillin, cefaclor, cefadroxil, cefamandole, cefazoline, cefdinir, cefepime, cefixime, cefinenoxime, cefinetazole, cefodizime, cefonicid, cefoperazone, ceforanide, cefotaxime, cefotetan, cefotiam, cefoxitin, cefpiramide, cefpodoxime, cefprozil, cefsulodin, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefuroxime, cephacetrile, cephalexin, cephaloglycin, cephaloridine, cephalothin, cephapirin, cephradine, chloramphenicol, chlortetracycline, cilastatin, cinnamycin, ciprofloxacin, clarithromycin, clavulanic acid, clindamycin, clioquinol, cloxacillin, colistimethate, colistin, cyclacillin, cycloserine, cyclosporine, cyclo-(Leu-Pro), dactinomycin, dalbavancin, dalfopristin, daptomycin, daunorubicin, demeclocycline, detorubicin, dicloxacillin, dihydrostreptomycin, dirithromycin, doxorubicin, doxycycline, epirubicin, erythromycin, eveminomycin, floxacillin, fosfomycin, fusidic acid, gemifloxacin, gentamycin, gramicidin, griseofulvin, hetacillin, idarubicin, imipenem, iseganan, ivermectin, kanamycin, laspartomycin, linezolid, linocomycin, loracarbef, magainin, meclocycline, meropenem, methacycline, methicillin, mezlocillin, minocycline, mitomycin, moenomycin, moxalactam, moxifloxacin, mycophenolic acid, nafcillin, natamycin, neomycin, netilmicin, niphimycin, nitrofurantoin, novobiocin, oleandomycin, oritavancin, oxacillin, oxytetracycline, paromomycin, penicillamine, penicillin G, penicillin V, phenethicillin, piperacillin, plicamycin, polymyxin B, pristinamycin, quinupristin, rifabutin, rifampin, rifamycin, rolitetracycline, sisomicin, spectrinomycin, streptomycin, streptozocin, sulbactam, sultamicillin, tacrolimus, tazobactam, teicoplanin, telithromycin, tetracycline, ticarcillin, tigecycline, tobramycin, troleandomycin, tunicamycin, tyrthricin, vancomycin, vidarabine, viomycin, virginiamycin, BMS-284,756, L-749,345, ER-35,786, S-4661, L-786,392, MC-02479, Pep5, RP 59500, and TD-6424. In some embodiments, two or more combined agents (e.g., a composition comprising a nanoemulsion and an antibiotic) may be used together or sequentially. In some embodiments, an antibiotic may comprise bacteriocins, type A lantibiotics, type B lantibiotics, liposidomycins, mureidomycins, alanoylcholines, quinolines, eveminomycins, glycylcyclines, carbapenems, cephalosporins, streptogramins, oxazolidonones, tetracyclines, cyclothialidines, bioxalomycins, cationic peptides, and/or protegrins. In some embodiments, the composition comprises lysostaphin.
  • The present invention is not limited by the type of nanoemulsion utilized (e.g., for respiratory administration, administration to a burn wound and/or for use in an immunogenic composition for induction of protective immune responses). Indeed, a variety of nanoemulsion compositions are contemplated to be useful in the present invention.
  • For example, in some embodiments, a nanoemulsion comprises (i) an aqueous phase; (ii) an oil phase; and at least one additional compound. In some embodiments of the present invention, these additional compounds are admixed into either the aqueous or oil phases of the composition. In other embodiments, these additional compounds are admixed into a composition of previously emulsified oil and aqueous phases. In certain of these embodiments, one or more additional compounds are admixed into an existing emulsion composition immediately prior to its use. In other embodiments, one or more additional compounds are admixed into an existing emulsion composition prior to the compositions immediate use.
  • Additional compounds suitable for use in a nanoemulsion of the present invention include, but are not limited to, one or more organic, and more particularly, organic phosphate based solvents, surfactants and detergents, cationic halogen containing compounds, germination enhancers, interaction enhancers, food additives (e.g., flavorings, sweeteners, bulking agents, and the like) and pharmaceutically acceptable compounds. Certain exemplary embodiments of the various compounds contemplated for use in the compositions of the present invention are presented below. Unless described otherwise, nanoemulsions are described in undiluted form.
  • Stability on Storage and after Application of the Nanoemulsions of the Invention
  • Storage Stability
  • The nanoemulsions of the invention can be stable at about 40° C. and about 75% relative humidity for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, or at least up to about 3 years.
  • In another embodiment of the invention, the nanoemulsions of the invention can be stable at about 25° C. and about 60% relative humidity for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, or at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, or at least up to about 5 years.
  • Further, the nanoemulsions of the invention can be stable at about 4° C. for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, at least up to about 5 years, at least up to about 5.5 years, at least up to about 6 years, at least up to about 6.5 years, or at least up to about 7 years.
  • Stability Upon Application
  • The nanoemulsions of the invention are stable upon application, as surprisingly the nanoemulsions do not lose their physical structure upon application. Microscopic examination of skin surface following application of a nanoemulsion according to the invention demonstrates the physical integrity of the nanoemulsions of the invention. This physical integrity may result in the desired absorption observed with the nanoemulsions of the invention.
  • Nanoemulsions
  • The term “nanoemulsion”, as defined herein, refers to a dispersion or droplet or any other lipid structure. Typical lipid structures contemplated in the invention include, but are not limited to, unilamellar, paucilamellar and multilamellar lipid vesicles, micelles and lamellar phases.
  • The nanoemulsion of the present invention comprises droplets having an average diameter size of less than about 1,000 nm, less than about 950 nm, less than about 900 nm, less than about 850 nm, less than about 800 nm, less than about 750 nm, less than about 700 nm, less than about 650 nm, less than about 600 nm, less than about 550 nm, less than about 500 nm, less than about 450 nm, less than about 400 nm, less than about 350 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, or any combination thereof. In one embodiment, the droplets have an average diameter size greater than about 125 nm and less than or equal to about 300 nm. In a different embodiment, the droplets have an average diameter size greater than about 50 nm or greater than about 70 nm, and less than or equal to about 125 nm. In other embodiments of the invention, the nanoemulsion droplets have an average diameter of from about 300 nm to about 600 nm; or the nanoemulsion droplets have an average diameter of from about 150 nm to about 400 nm.
  • In one embodiment of the invention, the nanoemulsion has a narrow range of MIC (minimum inhibitory concentration) and MBC (minimum bactericidal concentrations) values. In another embodiment, the MIC and MBC for the nanoemulsion differ by less than or equal to four-fold, meaning that the nanoemulsion is bactericidal. In addition, the MIC and MBC for the nanoemulsion may differ by greater than four-fold, meaning that the nanoemulsion is bacteriostatic.
  • In one embodiment of the invention, the nanoemulsion comprises: (a) an aqueous phase; (b) about 1% oil to about 80% oil; (c) about 0.1% organic solvent to about 50% organic solvent; (d) about 0.001% surfactant or detergent to about 10% surfactant or detergent; (e) about 0.0005% to about 1.0% of a chelating agent; or (e) any combination thereof. In another embodiment of the invention, the nanoemulsion comprises: (a) about 10% oil to about 80% oil; (b) about 1% organic solvent to about 50% organic solvent; (c) at least one non-ionic surfactant present in an amount of about 0.1% to about 10%; (d) at least one cationic agent present in an amount of about 0.01% to about 3%; or any combination thereof.
  • In another embodiment, the nanoemulsion comprises a cationic surfactant which is either cetylpyridinium chloride (CPC) or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or combination thereof. The cationic surfactant may have a concentration in the nanoemulsion of less than about 5.0% and greater than about 0.001%, or further, may have a concentration of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, less than about 0.10%, greater than about 0.001%, greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, and greater than about 0.010%.
  • In a further embodiment, the nanoemulsion comprises a non-ionic surfactant, and may have a concentration of about 0.01% to about 10.0%, or about 0.1% to about 3% of a non-ionic surfactant, such as a polysorbate.
  • In yet other embodiments of the invention, the nanoemulsion: (a) comprises at least one cationic surfactant; (b) comprises a cationic surfactant which is either cetylpyridinium chloride or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or combination thereof; (c) comprises a cationic surfactant, and wherein the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001%; (d) comprises a cationic surfactant, and wherein the concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, less than about 0.10%, greater than about 0.001%, greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, and greater than about 0.010%; or (e) any combination thereof. In yet other embodiments, (a) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant; (b) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant; (c) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a polysorbate nonionic surfactant; (d) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant, and the non-ionic surfactant is present in a concentration of about 0.05% to about 10%, about 0.05% to about 7.0%, about 0.1% to about 7%, or about 0.5% to about 5%; (e) the nanoemulsion comprises at least one cationic surfactant and at least one a nonionic surfactant, wherein the cationic surfactant is present in a concentration of about 0.05% to about 2% or about 0.01% to about 2%; or (f) any combination thereof.
  • In other embodiments, the nanoemulsion comprises: (a) water; (b) ethanol or glycerol (glycerine), or a combination thereof; (c) either cetylpyridinium chloride (CPC), or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or a combination thereof; (c) soybean oil; and (e) Poloxamer 407, Tween 80, or Tween 20. The nanoemulsion can further comprise EDTA.
  • These quantities of each component present in the nanoemulsion refer to a therapeutic nanoemulsion, and not to a nanoemulsion to be tested in vitro. This is significant, as nanoemulsions tested in vitro generally have lower concentrations of oil, organic solvent, surfactant or detergent, and (if present) chelating agent than that present in a nanoemulsion intended for therapeutic use, e.g., topical use. This is because in vitro studies do not require the nanoemulsion droplets to traverse the skin. For topical, aerosol, intradermal etc. use, the concentrations of the components must be higher to result in a therapeutic nanoemulsion. However, the relative quantities of each component used in a nanoemulsion tested in vitro are applicable to a nanoemulsion to be used therapeutically and, therefore, in vitro quantities can be scaled up to prepare a therapeutic composition, and in vitro data is predictive of topical application success.
  • 1. Aqueous Phase
  • The aqueous phase can comprise any type of aqueous phase including, but not limited to, water (e.g., H2O, distilled water, tap water) and solutions (e.g., phosphate buffered saline (PBS) solution). In certain embodiments, the aqueous phase comprises water at a pH of about 4 to 10, preferably about 6 to 8. The water can be deionized (hereinafter “DiH2O”). In some embodiments the aqueous phase comprises phosphate buffered saline (PBS). The aqueous phase may further be sterile and pyrogen free.
  • 2. Organic Solvents
  • Organic solvents in the nanoemulsions of the invention include, but are not limited to, C1-C12 alcohol, diol, triol, dialkyl phosphate, tri-alkyl phosphate, such as tri-n-butyl phosphate, semi-synthetic derivatives thereof, and combinations thereof. In one aspect of the invention, the organic solvent is an alcohol chosen from a nonpolar solvent, a polar solvent, a protic solvent, or an aprotic solvent.
  • Suitable organic solvents for the nanoemulsion include, but are not limited to, ethanol, methanol, isopropyl alcohol, glycerol, medium chain triglycerides, diethyl ether, ethyl acetate, acetone, dimethyl sulfoxide (DMSO), acetic acid, n-butanol, butylene glycol, perfumers alcohols, isopropanol, n-propanol, formic acid, propylene glycols, glycerol, sorbitol, industrial methylated spirit, triacetin, hexane, benzene, toluene, diethyl ether, chloroform, 1,4-dixoane, tetrahydrofuran, dichloromethane, acetone, acetonitrile, dimethylformamide, dimethyl sulfoxide, formic acid, semi-synthetic derivatives thereof, and any combination thereof.
  • 3. Oil Phase
  • The oil in the nanoemulsion of the invention can be any cosmetically or pharmaceutically acceptable oil. The oil can be volatile or non-volatile, and may be chosen from animal oil, vegetable oil, natural oil, synthetic oil, hydrocarbon oils, silicone oils, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable oils include, but are not limited to, mineral oil, squalene oil, flavor oils, silicon oil, essential oils, water insoluble vitamins, Isopropyl stearate, Butyl stearate, Octyl palmitate, Cetyl palmitate, Tridecyl behenate, Diisopropyl adipate, Dioctyl sebacate, Menthyl anthranhilate, Cetyl octanoate, Octyl salicylate, Isopropyl myristate, neopentyl glycol dicarpate cetols, Ceraphyls®, Decyl oleate, diisopropyl adipate, C12-15 alkyl lactates, Cetyl lactate, Lauryl lactate, Isostearyl neopentanoate, Myristyl lactate, Isocetyl stearoyl stearate, Octyldodecyl stearoyl stearate, Hydrocarbon oils, Isoparaffin, Fluid paraffins, Isododecane, Petrolatum, Argan oil, Canola oil, Chile oil, Coconut oil, corn oil, Cottonseed oil, Flaxseed oil, Grape seed oil, Mustard oil, Olive oil, Palm oil, Palm kernel oil, Peanut oil, Pine seed oil, Poppy seed oil, Pumpkin seed oil, Rice bran oil, Safflower oil, Tea oil, Truffle oil, Vegetable oil, Apricot (kernel) oil, Jojoba oil (simmondsia chinensis seed oil), Grapeseed oil, Macadamia oil, Wheat germ oil, Almond oil, Rapeseed oil, Gourd oil, Soybean oil, Sesame oil, Hazelnut oil, Maize oil, Sunflower oil, Hemp oil, Bois oil, Kuki nut oil, Avocado oil, Walnut oil, Fish oil, berry oil, allspice oil, juniper oil, seed oil, almond seed oil, anise seed oil, celery seed oil, cumin seed oil, nutmeg seed oil, leaf oil, basil leaf oil, bay leaf oil, cinnamon leaf oil, common sage leaf oil, eucalyptus leaf oil, lemon grass leaf oil, melaleuca leaf oil, oregano leaf oil, patchouli leaf oil, peppermint leaf oil, pine needle oil, rosemary leaf oil, spearmint leaf oil, tea tree leaf oil, thyme leaf oil, wintergreen leaf oil, flower oil, chamomile oil, clary sage oil, clove oil, geranium flower oil, hyssop flower oil, jasmine flower oil, lavender flower oil, manuka flower oil, Marhoram flower oil, orange flower oil, rose flower oil, ylang-ylang flower oil, Bark oil, cassia Bark oil, cinnamon bark oil, sassafras Bark oil, Wood oil, camphor wood oil, cedar wood oil, rosewood oil, sandalwood oil), rhizome (ginger) wood oil, resin oil, frankincense oil, myrrh oil, peel oil, bergamot peel oil, grapefruit peel oil, lemon peel oil, lime peel oil, orange peel oil, tangerine peel oil, root oil, valerian oil, Oleic acid, Linoleic acid, Oleyl alcohol, Isostearyl alcohol, semi-synthetic derivatives thereof, and any combinations thereof.
  • The oil may further comprise a silicone component, such as a volatile silicone component, which can be the sole oil in the silicone component or can be combined with other silicone and non-silicone, volatile and non-volatile oils. Suitable silicone components include, but are not limited to, methylphenylpolysiloxane, simethicone, dimethicone, phenyltrimethicone (or an organomodified version thereof), alkylated derivatives of polymeric silicones, cetyl dimethicone, lauryl trimethicone, hydroxylated derivatives of polymeric silicones, such as dimethiconol, volatile silicone oils, cyclic and linear silicones, cyclomethicone, derivatives of cyclomethicone, hexamethylcyclotrisiloxane, octamethylcyclotetrasiloxane, decamethylcyclopentasiloxane, volatile linear dimethylpolysiloxanes, isohexadecane, isoeicosane, isotetracosane, polyisobutene, isooctane, isododecane, semi-synthetic derivatives thereof, and combinations thereof.
  • The volatile oil can be the organic solvent, or the volatile oil can be present in addition to an organic solvent. Suitable volatile oils include, but are not limited to, a terpene, monoterpene, sesquiterpene, carminative, azulene, menthol, camphor, thujone, thymol, nerol, linalool, limonene, geraniol, perillyl alcohol, nerolidol, farnesol, ylangene, bisabolol, farnesene, ascaridole, chenopodium oil, citronellal, citral, citronellol, chamazulene, yarrow, guaiazulene, chamomile, semi-synthetic derivatives, or combinations thereof.
  • In one aspect of the invention, the volatile oil in the silicone component is different than the oil in the oil phase.
  • 4. Surfactants/Detergents
  • The surfactant or detergent in the nanoemulsion of the invention can be a pharmaceutically acceptable ionic surfactant, a pharmaceutically acceptable nonionic surfactant, a pharmaceutically acceptable cationic surfactant, a pharmaceutically acceptable anionic surfactant, or a pharmaceutically acceptable zwitterionic surfactant.
  • Exemplary useful surfactants are described in Applied Surfactants: Principles and Applications. Tharwat F. Tadros, Copyright 8 2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-30629-3), which is specifically incorporated by reference.
  • Further, the surfactant can be a pharmaceutically acceptable ionic polymeric surfactant, a pharmaceutically acceptable nonionic polymeric surfactant, a pharmaceutically acceptable cationic polymeric surfactant, a pharmaceutically acceptable anionic polymeric surfactant, or a pharmaceutically acceptable zwitterionic polymeric surfactant. Examples of polymeric surfactants include, but are not limited to, a graft copolymer of a poly(methyl methacrylate) backbone with multiple (at least one) polyethylene oxide (PEO) side chain, polyhydroxystearic acid, an alkoxylated alkyl phenol formaldehyde condensate, a polyalkylene glycol modified polyester with fatty acid hydrophobes, a polyester, semi-synthetic derivatives thereof, or combinations thereof.
  • Surface active agents or surfactants, are amphipathic molecules that consist of a non-polar hydrophobic portion, usually a straight or branched hydrocarbon or fluorocarbon chain containing 8-18 carbon atoms, attached to a polar or ionic hydrophilic portion. The hydrophilic portion can be nonionic, ionic or zwitterionic. The hydrocarbon chain interacts weakly with the water molecules in an aqueous environment, whereas the polar or ionic head group interacts strongly with water molecules via dipole or iondipole interactions. Based on the nature of the hydrophilic group, surfactants are classified into anionic, cationic, zwitterionic, nonionic and polymeric surfactants.
  • Suitable surfactants include, but are not limited to, ethoxylated nonylphenol comprising 9 to 10 units of ethyleneglycol, ethoxylated undecanol comprising 8 units of ethyleneglycol, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate, polyoxyethylene (20) sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monostearate, sorbitan monooleate, ethoxylated hydrogenated ricin oils, sodium laurylsulfate, a diblock copolymer of ethyleneoxyde and propyleneoxyde, Ethylene Oxide-Propylene Oxide Block Copolymers, and tetra-functional block copolymers based on ethylene oxide and propylene oxide, Glyceryl monoesters, Glyceryl caprate, Glyceryl caprylate, Glyceryl cocate, Glyceryl erucate, Glyceryl hydroxysterate, Glyceryl isostearate, Glyceryl lanolate, Glyceryl laurate, Glyceryl linolate, Glyceryl myristate, Glyceryl oleate, Glyceryl PABA, Glyceryl palmitate, Glyceryl ricinoleate, Glyceryl stearate, Glyceryl thighlycolate, Glyceryl dilaurate, Glyceryl dioleate, Glyceryl dimyristate, Glyceryl disterate, Glyceryl sesuioleate, Glyceryl stearate lactate, Polyoxyethylene cetyl/stearyl ether, Polyoxyethylene cholesterol ether, Polyoxyethylene laurate or dilaurate, Polyoxyethylene stearate or distearate, polyoxyethylene fatty ethers, Polyoxyethylene lauryl ether, Polyoxyethylene stearyl ether, polyoxyethylene myristyl ether, a steroid, Cholesterol, Betasitosterol, Bisabolol, fatty acid esters of alcohols, isopropyl myristate, Aliphati-isopropyl n-butyrate, Isopropyl n-hexanoate, Isopropyl n-decanoate, Isoproppyl palmitate, Octyldodecyl myristate, alkoxylated alcohols, alkoxylated acids, alkoxylated amides, alkoxylated sugar derivatives, alkoxylated derivatives of natural oils and waxes, polyoxyethylene polyoxypropylene block copolymers, nonoxynol-14, PEG-8 laurate, PEG-6 Cocoamide, PEG-20 methylglucose sesquistearate, PEG40 lanolin, PEG-40 castor oil, PEG-40 hydrogenated castor oil, polyoxyethylene fatty ethers, glyceryl diesters, polyoxyethylene stearyl ether, polyoxyethylene myristyl ether, and polyoxyethylene lauryl ether, glyceryl dilaurate, glyceryl dimystate, glyceryl distearate, semi-synthetic derivatives thereof, or mixtures thereof.
  • Additional suitable surfactants include, but are not limited to, non-ionic lipids, such as glyceryl laurate, glyceryl myristate, glyceryl dilaurate, glyceryl dimyristate, semi-synthetic derivatives thereof, and mixtures thereof.
  • In additional embodiments, the surfactant is a polyoxyethylene fatty ether having a polyoxyethylene head group ranging from about 2 to about 100 groups, or an alkoxylated alcohol having the structure R5—(OCH2CH2)y—OH, wherein R5 is a branched or unbranched alkyl group having from about 6 to about 22 carbon atoms and y is between about 4 and about 100, and preferably, between about 10 and about 100. Preferably, the alkoxylated alcohol is the species wherein R5 is a lauryl group and y has an average value of 23.
  • In a different embodiment, the surfactant is an alkoxylated alcohol which is an ethoxylated derivative of lanolin alcohol. Preferably, the ethoxylated derivative of lanolin alcohol is laneth-10, which is the polyethylene glycol ether of lanolin alcohol with an average ethoxylation value of 10.
  • Nonionic surfactants include, but are not limited to, an ethoxylated surfactant, an alcohol ethoxylated, an alkyl phenol ethoxylated, a fatty acid ethoxylated, a monoalkaolamide ethoxylated, a sorbitan ester ethoxylated, a fatty amino ethoxylated, an ethylene oxide-propylene oxide copolymer, Bis(polyethylene glycol bis[imidazoyl carbonyl]), nonoxynol-9, Bis(polyethylene glycol bis[imidazoyl carbonyl]), Brij® 35, Brij® 56, Brij® 72, Brij® 76, Brij® 92V, Brij® 97, Brij® 58P, Cremophor® EL, Decaethylene glycol monododecyl ether, N-Decanoyl-N-methylglucamine, n-Decyl alpha-D-glucopyranoside, Decyl beta-D-maltopyranoside, n-Dodecanoyl-N-methylglucamide, n-Dodecyl alpha-D-maltoside, n-Dodecyl beta-D-maltoside, n-Dodecyl beta-D-maltoside, Heptaethylene glycol monodecyl ether, Heptaethylene glycol monododecyl ether, Heptaethylene glycol monotetradecyl ether, n-Hexadecyl beta-D-maltoside, Hexaethylene glycol monododecyl ether, Hexaethylene glycol monohexadecyl ether, Hexaethylene glycol monooctadecyl ether, Hexaethylene glycol monotetradecyl ether, Igepal CA-630, Igepal CA-630, Methyl-6-O-(N-heptylcarbamoyl)-alpha-D-glucopyranoside, Nonaethylene glycol monododecyl ether, N—N-Nonanoyl-N-methylglucamine, Octaethylene glycol monodecyl ether, Octaethylene glycol monododecyl ether, Octaethylene glycol monohexadecyl ether, Octaethylene glycol monooctadecyl ether, Octaethylene glycol monotetradecyl ether, Octyl-beta-D-glucopyranoside, Pentaethylene glycol monodecyl ether, Pentaethylene glycol monododecyl ether, Pentaethylene glycol monohexadecyl ether, Pentaethylene glycol monohexyl ether, Pentaethylene glycol monooctadecyl ether, Pentaethylene glycol monooctyl ether, Polyethylene glycol diglycidyl ether, Polyethylene glycol ether W-1, Polyoxyethylene 10 tridecyl ether, Polyoxyethylene 100 stearate, Polyoxyethylene 20 isohexadecyl ether, Polyoxyethylene 20 oleyl ether, Polyoxyethylene 40 stearate, Polyoxyethylene 50 stearate, Polyoxyethylene 8 stearate, Polyoxyethylene bis(imidazolyl carbonyl), Polyoxyethylene 25 propylene glycol stearate, Saponin from Quillaja bark, Span® 20, Span® 40, Span® 60, Span® 65, Span® 80, Span® 85, Tergitol, Type 15-S-12, Tergitol, Type 15-S-30, Tergitol, Type 15-S-5, Tergitol, Type 15-S-7, Tergitol, Type 15-S-9, Tergitol, Type NP-10, Tergitol, Type NP-4, Tergitol, Type NP-40, Tergitol, Type NP-7, Tergitol, Type NP-9, Tergitol, Tergitol, Type TMN-10, Tergitol, Type TMN-6, Tetradecyl-beta-D-maltoside, Tetraethylene glycol monodecyl ether, Tetraethylene glycol monododecyl ether, Tetraethylene glycol monotetradecyl ether, Triethylene glycol monodecyl ether, Triethylene glycol monododecyl ether, Triethylene glycol monohexadecyl ether, Triethylene glycol monooctyl ether, Triethylene glycol monotetradecyl ether, Triton CF-21, Triton CF-32, Triton DF-12, Triton DF-16, Triton GR-5M, Triton QS-15, Triton QS-44, Triton X-100, Triton X-102, Triton X-15, Triton X-151, Triton X-200, Triton X-207, Triton® X-114, Triton® X-165, Triton® X-305, Triton® X-405, Triton® X-45, Triton® X-705-70, TWEEN® 20, TWEEN® 21, TWEEN® 40, TWEEN® 60, TWEEN® 61, TWEEN® 65, TWEEN® 80, TWEEN® 81, TWEEN® 85, Tyloxapol, n-Undecyl beta-D-glucopyranoside, semi-synthetic derivatives thereof, or combinations thereof.
  • In addition, the nonionic surfactant can be a poloxamer. Poloxamers are polymers made of a block of polyoxyethylene, followed by a block of polyoxypropylene, followed by a block of polyoxyethylene. The average number of units of polyoxyethylene and polyoxypropylene varies based on the number associated with the polymer. For example, the smallest polymer, Poloxamer 101, consists of a block with an average of 2 units of polyoxyethylene, a block with an average of 16 units of polyoxypropylene, followed by a block with an average of 2 units of polyoxyethylene. Poloxamers range from colorless liquids and pastes to white solids. In cosmetics and personal care products, Poloxamers are used in the formulation of skin cleansers, bath products, shampoos, hair conditioners, mouthwashes, eye makeup remover and other skin and hair products. Examples of Poloxamers include, but are not limited to, Poloxamer 101, Poloxamer 105, Poloxamer 108, Poloxamer 122, Poloxamer 123, Poloxamer 124, Poloxamer 181, Poloxamer 182, Poloxamer 183, Poloxamer 184, Poloxamer 185, Poloxamer 188, Poloxamer 212, Poloxamer 215, Poloxamer 217, Poloxamer 231, Poloxamer 234, Poloxamer 235, Poloxamer 237, Poloxamer 238, Poloxamer 282, Poloxamer 284, Poloxamer 288, Poloxamer 331, Poloxamer 333, Poloxamer 334, Poloxamer 335, Poloxamer 338, Poloxamer 401, Poloxamer 402, Poloxamer 403, Poloxamer 407, Poloxamer 105 Benzoate, and Poloxamer 182 Dibenzoate.
  • Suitable cationic surfactants include, but are not limited to, a quarternary ammonium compound, an alkyl trimethyl ammonium chloride compound, a dialkyl dimethyl ammonium chloride compound, a cationic halogen-containing compound, such as cetylpyridinium chloride, Benzalkonium chloride, Benzalkonium chloride, Benzyldimethylhexadecylammonium chloride, Benzyldimethyltetradecylammonium chloride, Benzyldodecyldimethylammonium bromide, Benzyltrimethylammonium tetrachloroiodate, Dimethyldioctadecylammonium bromide, Dodecylethyldimethylammonium bromide, Dodecyltrimethylammonium bromide, Dodecyltrimethylammonium bromide, Ethylhexadecyldimethylammonium bromide, Girard's reagent T, Hexadecyltrimethylammonium bromide, Hexadecyltrimethylammonium bromide, N,N′,N′-Polyoxyethylene(10)-N-tallow-1,3-diaminopropane, Thonzonium bromide, Trimethyl(tetradecyl)ammonium bromide, 1,3,5-Triazine-1,3,5(2H,4H,6H)-triethanol, 1-Decanaminium, N-decyl-N,N-dimethyl-, chloride, Didecyl dimethyl ammonium chloride, 2-(2-(p-(Diisobutyl)cresosxy)ethoxy)ethyl dimethyl benzyl ammonium chloride, 2-(2-(p-(Diisobutyl)phenoxy)ethoxy)ethyl dimethyl benzyl ammonium chloride, Alkyl 1 or 3 benzyl-1-(2-hydroxethyl)-2-imidazolinium chloride, Alkyl bis(2-hydroxyethyl)benzyl ammonium chloride, Alkyl demethyl benzyl ammonium chloride, Alkyl dimethyl 3,4-dichlorobenzyl ammonium chloride (100% C12), Alkyl dimethyl 3,4-dichlorobenzyl ammonium chloride (50% C14, 40% C12, 10% C16), Alkyl dimethyl 3,4-dichlorobenzyl ammonium chloride (55% C14, 23% C12, 20% C16), Alkyl dimethyl benzyl ammonium chloride, Alkyl dimethyl benzyl ammonium chloride (100% C14), Alkyl dimethyl benzyl ammonium chloride (100% C16), Alkyl dimethyl benzyl ammonium chloride (41% C14, 28% C12), Alkyl dimethyl benzyl ammonium chloride (47% C12, 18% C14), Alkyl dimethyl benzyl ammonium chloride (55% C16, 20% C14), Alkyl dimethyl benzyl ammonium chloride (58% C14, 28% C16), Alkyl dimethyl benzyl ammonium chloride (60% C14, 25% C12), Alkyl dimethyl benzyl ammonium chloride (61% C11, 23% C14), Alkyl dimethyl benzyl ammonium chloride (61% C12, 23% C14), Alkyl dimethyl benzyl ammonium chloride (65% C12, 25% C14), Alkyl dimethyl benzyl ammonium chloride (67% C12, 24% C14), Alkyl dimethyl benzyl ammonium chloride (67% C12, 25% C14), Alkyl dimethyl benzyl ammonium chloride (90% C14, 5% C12), Alkyl dimethyl benzyl ammonium chloride (93% C14, 4% C12), Alkyl dimethyl benzyl ammonium chloride (95% C16, 5% C18), Alkyl didecyl dimethyl ammonium chloride, Alkyl dimethyl benzyl ammonium chloride (C12-16), Alkyl dimethyl benzyl ammonium chloride (C12-18), dialkyl dimethyl benzyl ammonium chloride, Alkyl dimethyl dimethybenzyl ammonium chloride, Alkyl dimethyl ethyl ammonium bromide (90% C14, 5% C16, 5% C12), Alkyl dimethyl ethyl ammonium bromide (mixed alkyl and alkenyl groups as in the fatty acids of soybean oil), Alkyl dimethyl ethylbenzyl ammonium chloride, Alkyl dimethyl ethylbenzyl ammonium chloride (60% C14), Alkyl dimethyl isopropylbenzyl ammonium chloride (50% C12, 30% C14, 17% C16, 3% C18), Alkyl trimethyl ammonium chloride (58% C18, 40% C16, 1% C14, 1% C12), Alkyl trimethyl ammonium chloride (90% C18, 10% C16), Alkyldimethyl(ethylbenzyl) ammonium chloride (C12-18), Di-(C8-10)-alkyl dimethyl ammonium chlorides, Dialkyl dimethyl ammonium chloride, Dialkyl methyl benzyl ammonium chloride, Didecyl dimethyl ammonium chloride, Diisodecyl dimethyl ammonium chloride, Dioctyl dimethyl ammonium chloride, Dodecyl bis(2-hydroxyethyl) octyl hydrogen ammonium chloride, Dodecyl dimethyl benzyl ammonium chloride, Dodecylcarbamoyl methyl dimethyl benzyl ammonium chloride, Heptadecyl hydroxyethylimidazolinium chloride, Hexahydro-1,3,5-tris(2-hydroxyethyl)-s-triazine, Myristalkonium chloride (and) Quat RNIUM 14, N,N-Dimethyl-2-hydroxypropylammonium chloride polymer, n-Tetradecyl dimethyl benzyl ammonium chloride monohydrate, Octyl decyl dimethyl ammonium chloride, Octyl dodecyl dimethyl ammonium chloride, Octyphenoxyethoxyethyl dimethyl benzyl ammonium chloride, Oxydiethylenebis(alkyl dimethyl ammonium chloride), Trimethoxysily propyl dimethyl octadecyl ammonium chloride, Trimethoxysilyl quats, Trimethyl dodecylbenzyl ammonium chloride, semi-synthetic derivatives thereof, and combinations thereof.
  • Exemplary cationic halogen-containing compounds include, but are not limited to, cetylpyridinium halides, cetyltrimethylammonium halides, cetyldimethylethylammonium halides, cetyldimethylbenzylammonium halides, cetyltributylphosphonium halides, dodecyltrimethylammonium halides, or tetradecyltrimethylammonium halides. In some particular embodiments, suitable cationic halogen containing compounds comprise, but are not limited to, cetylpyridinium chloride (CPC), cetyltrimethylammonium chloride, cetylbenzyldimethylammonium chloride, cetylpyridinium bromide (CPB), cetyltrimethylammonium bromide (CTAB), cetyidimethylethylammonium bromide, cetyltributylphosphonium bromide, dodecyltrimethylammonium bromide, and tetrad ecyltrimethylammonium bromide. In particularly preferred embodiments, the cationic halogen containing compound is CPC, although the compositions of the present invention are not limited to formulation with a particular cationic containing compound.
  • Suitable anionic surfactants include, but are not limited to, a carboxylate, a sulphate, a sulphonate, a phosphate, chenodeoxycholic acid, chenodeoxycholic acid sodium salt, cholic acid, ox or sheep bile, Dehydrocholic acid, Deoxycholic acid, Deoxycholic acid, Deoxycholic acid methyl ester, Digitonin, Digitoxigenin, N,N-Dimethyldodecylamine N-oxide, Docusate sodium salt, Glycochenodeoxycholic acid sodium salt, Glycocholic acid hydrate, synthetic, Glycocholic acid sodium salt hydrate, synthetic, Glycodeoxycholic acid monohydrate, Glycodeoxycholic acid sodium salt, Glycolithocholic acid 3-sulfate disodium salt, Glycolithocholic acid ethyl ester, N-Lauroylsarcosine sodium salt, N-Lauroylsarcosine solution, N-Lauroylsarcosine solution, Lithium dodecyl sulfate, Lithium dodecyl sulfate, Lithium dodecyl sulfate, Lugol solution, Niaproof 4, Type 4,1-Octanesulfonic acid sodium salt, Sodium 1-butanesulfonate, Sodium 1-decanesulfonate, Sodium 1-decanesulfonate, Sodium 1-dodecanesulfonate, Sodium 1-heptanesulfonate anhydrous, Sodium 1-heptanesulfonate anhydrous, Sodium 1-nonanesulfonate, Sodium 1-propanesulfonate monohydrate, Sodium 2-bromoethanesulfonate, Sodium cholate hydrate, Sodium choleate, Sodium deoxycholate, Sodium deoxycholate monohydrate, Sodium dodecyl sulfate, Sodium hexanesulfonate anhydrous, Sodium octyl sulfate, Sodium pentanesulfonate anhydrous, Sodium taurocholate, Taurochenodeoxycholic acid sodium salt, Taurodeoxycholic acid sodium salt monohydrate, Taurohyodeoxycholic acid sodium salt hydrate, Taurolithocholic acid 3-sulfate disodium salt, Tauroursodeoxycholic acid sodium salt, Trizma® dodecyl sulfate, TWEEN® 80, Ursodeoxycholic acid, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable zwitterionic surfactants include, but are not limited to, an N-alkyl betaine, lauryl amindo propyl dimethyl betaine, an alkyl dimethyl glycinate, an N-alkyl amino propionate, CHAPS, minimum 98% (TLC), CHAPS, SigmaUltra, minimum 98% (TLC), CHAPS, for electrophoresis, minimum 98% (TLC), CHAPSO, minimum 98%, CHAPSO, SigmaUltra, CHAPSO, for electrophoresis, 3-(Decyldimethylammonio)propanesulfonate inner salt, 3-Dodecyldimethylammonio)propanesulfonate inner salt, SigmaUltra, 3-(Dodecyldimethylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylmyristylammonio)propanesulfonate, 3-(N,N-Dimethyloctadecylammonio)propanesulfonate, 3-(N,N-Dimethyloctylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylpalmitylammonio)propanesulfonate, semi-synthetic derivatives thereof, and combinations thereof.
  • In some embodiments, the nanoemulsion comprises a cationic surfactant, which can be cetylpyridinium chloride. In other embodiments of the invention, the nanoemulsion comprises a cationic surfactant, and the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001%. In yet another embodiment of the invention, the nanoemulsion comprises a cationic surfactant, and the concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, or less than about 0.10%. Further, the concentration of the cationic agent in the nanoemulsion is greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, greater than about 0.010%, or greater than about 0.001%. In one embodiment, the concentration of the cationic agent in the nanoemulsion is less than about 5.0% and greater than about 0.001%.
  • In another embodiment of the invention, the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant. The non-cationic surfactant is a nonionic surfactant, such as a polysorbate (Tween), such as polysorbate 80 or polysorbate 20. In one embodiment, the non-ionic surfactant is present in a concentration of about 0.05% to about 7.0%, or the non-ionic surfactant is present in a concentration of about 0.5% to about 4%. In yet another embodiment of the invention, the nanoemulsion comprises a cationic surfactant present in a concentration of about 0.01% to about 2%, in combination with a nonionic surfactant.
  • 5. Additional Ingredients
  • Additional compounds suitable for use in the nanoemulsions of the invention include but are not limited to one or more solvents, such as an organic phosphate-based solvent, bulking agents, coloring agents, pharmaceutically acceptable excipients, a preservative, pH adjuster, buffer, chelating agent, etc. The additional compounds can be admixed into a previously emulsified nanoemulsion, or the additional compounds can be added to the original mixture to be emulsified. In certain of these embodiments, one or more additional compounds are admixed into an existing nanoemulsion composition immediately prior to its use.
  • Suitable preservatives in the nanoemulsions of the invention include, but are not limited to, cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha-tocophernol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, sodium ascorbate, sodium metabisulphite, citric acid, edetic acid, semi-synthetic derivatives thereof, and combinations thereof. Other suitable preservatives include, but are not limited to, benzyl alcohol, chlorhexidine (bis(p-chlorophenyldiguanido) hexane), chlorphenesin (3-(-4-chloropheoxy)-propane-1,2-diol), Kathon CG (methyl and methylchloroisothiazolinone), parabens (methyl, ethyl, propyl, butyl hydrobenzoates), phenoxyethanol (2-phenoxyethanol), sorbic acid (potassium sorbate, sorbic acid), Phenonip (phenoxyethanol, methyl, ethyl, butyl, propyl parabens), Phenoroc (phenoxyethanol 0.73%, methyl paraben 0.2%, propyl paraben 0.07%), Liquipar Oil (isopropyl, isobutyl, butylparabens), Liquipar PE (70% phenoxyethanol, 30% liquipar oil), Nipaguard MPA (benzyl alcohol (70%), methyl & propyl parabens), Nipaguard MPS (propylene glycol, methyl & propyl parabens), Nipasept (methyl, ethyl and propyl parabens), Nipastat (methyl, butyl, ethyl and propyel parabens), Elestab 388 (phenoxyethanol in propylene glycol plus chlorphenesin and methylparaben), and Killitol (7.5% chlorphenesin and 7.5% methyl parabens).
  • The nanoemulsion may further comprise at least one pH adjuster. Suitable pH adjusters in the nanoemulsion of the invention include, but are not limited to, diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof.
  • In addition, the nanoemulsion can comprise a chelating agent. In one embodiment of the invention, the chelating agent is present in an amount of about 0.0005% to about 1.0%. Examples of chelating agents include, but are not limited to, phytic acid, polyphosphoric acid, citric acid, gluconic acid, acetic acid, lactic acid, ethylenediamine, ethylenediaminetetraacetic acid (EDTA), and dimercaprol, and a preferred chelating agent is ethylenediaminetetraacetic acid.
  • The nanoemulsion can comprise a buffering agent, such as a pharmaceutically acceptable buffering agent. Examples of buffering agents include, but are not limited to, 2-Amino-2-methyl-1,3-propanediol, ≧9.5% (NT), 2-Amino-2-methyl-1-propanol, ≧99.0% (GC), L-(+)-Tartaric acid, ≧99.5% (T), ACES, ≧99.5% (T), ADA, ≧99.0% (T), Acetic acid, ≧09.5% (GC/T), Acetic acid, for luminescence, ≧99.5% (GC/T), Ammonium acetate solution, for molecular biology, ˜5 M in H2O, Ammonium acetate, for luminescence, ≧99.0% (calc. on dry substance, T), Ammonium bicarbonate, ≧99.5% (T), Ammonium citrate dibasic, ≧99.0% (T), Ammonium formate solution, 10 M in H2O, Ammonium formate, ≧99.0% (calc. based on dry substance, NT), Ammonium oxalate monohydrate, ≧99.5% (RT), Ammonium phosphate dibasic solution, 2.5 M in H2O, Ammonium phosphate dibasic, ≧99.0% (T), Ammonium phosphate monobasic solution, 2.5 M in H2O, Ammonium phosphate monobasic, ≧99.5% (T), Ammonium sodium phosphate dibasic tetrahydrate, ≧99.5% (NT), Ammonium sulfate solution, for molecular biology, 3.2 M in H2O, Ammonium tartrate dibasic solution, 2 M in H2O (colorless solution at 20° C.), Ammonium tartrate dibasic, ≧99.5% (T), BES buffered saline, for molecular biology, 2× concentrate, BES, ≧99.5% (T), BES, for molecular biology, ≧99.5% (T), BICINE buffer Solution, for molecular biology, 1 M in H2O, BICINE, ≧99.5% (T), BIS-TRIS, ≧99.0% (NT), Bicarbonate buffer solution, >0.1 M Na2CO3, >0.2 M NaHCO3, Boric acid, ≧99.5% (T), Boric acid, for molecular biology, ≧99.5% (T), CAPS, 99.0% (TLC), CHES, ≧99.5% (T), Calcium acetate hydrate, ≧99.0% (calc. on dried material, KT), Calcium carbonate, precipitated, ≧99.0% (KT), Calcium citrate tribasic tetrahydrate, ≧98.0% (calc. on dry substance, KT), Citrate Concentrated Solution, for molecular biology, 1 M in H2O, Citric acid, anhydrous, ≧99.5% (T), Citric acid, for luminescence, anhydrous, ≧99.5% (T), Diethanolamine, ≧99.5% (GC), EPPS, ≧99.0% (T), Ethylenediaminetetraacetic acid disodium salt dihydrate, for molecular biology, ≧99.0% (T), Formic acid solution, 1.0 M in H2O, Gly-Gly-Gly, ≧99.0% (NT), Gly-Gly, ≧99.5% (NT), Glycine, ≧99.0% (NT), Glycine, for luminescence, ≧99.0% (NT), Glycine, for molecular biology, ≧99.0% (NT), HEPES buffered saline, for molecular biology, 2× concentrate, HEPES, ≧99.5% (T), HEPES, for molecular biology, ≧99.5% (T), Imidazole buffer Solution, 1 M in H2O, Imidazole, ≧99.5% (GC), Imidazole, for luminescence, ≧99.5% (GC), Imidazole, for molecular biology, ≧99.5% (GC), Lipoprotein Refolding Buffer, Lithium acetate dihydrate, ≧99.0% (NT), Lithium citrate tribasic tetrahydrate, ≧99.5% (NT), MES hydrate, ≧99.5% (T), MES monohydrate, for luminescence, ≧99.5% (T), MES solution, for molecular biology, 0.5 M in H2O, MOPS, ≧99.5% (T), MOPS, for luminescence, ≧99.5% (T), MOPS, for molecular biology, ≧99.5% (T), Magnesium acetate solution, for molecular biology, ˜1 M in H2O, Magnesium acetate tetrahydrate, ≧99.0% (KT), Magnesium citrate tribasic nonahydrate, ≧98.0% (calc. based on dry substance, KT), Magnesium formate solution, 0.5 M in H2O, Magnesium phosphate dibasic trihydrate, ≧98.0% (KT), Neutralization solution for the in-situ hybridization for in-situ hybridization, for molecular biology, Oxalic acid dihydrate, ≧99.5% (RT), PIPES, ≧99.5% (T), PIPES, for molecular biology, ≧99.5% (T), Phosphate buffered saline, solution (autoclaved), Phosphate buffered saline, washing buffer for peroxidase conjugates in Western Blotting, 10× concentrate, piperazine, anhydrous, ≧99.0% (T), Potassium D-tartrate monobasic, ≧99.0% (T), Potassium acetate solution, for molecular biology, Potassium acetate solution, for molecular biology, 5 M in H2O, Potassium acetate solution, for molecular biology, ˜1 M in H2O, Potassium acetate, ≧99.0% (NT), Potassium acetate, for luminescence, ≧99.0% (NT), Potassium acetate, for molecular biology, ≧99.0% (NT), Potassium bicarbonate, ≧99.5% (T), Potassium carbonate, anhydrous, ≧99.0% (T), Potassium chloride, ≧99.5% (AT), Potassium citrate monobasic, ≧99.0% (dried material, NT), Potassium citrate tribasic solution, 1 M in H2O, Potassium formate solution, 14 M in H2O, Potassium formate, ≧99.5% (NT), Potassium oxalate monohydrate, ≧99.0% (RT), Potassium phosphate dibasic, anhydrous, ≧99.0% (T), Potassium phosphate dibasic, for luminescence, anhydrous, ≧99.0% (T), Potassium phosphate dibasic, for molecular biology, anhydrous, ≧99.0% (T), Potassium phosphate monobasic, anhydrous, ≧99.5% (T), Potassium phosphate monobasic, for molecular biology, anhydrous, ≧99.5% (T), Potassium phosphate tribasic monohydrate, ≧95% (T), Potassium phthalate monobasic, ≧99.5% (T), Potassium sodium tartrate solution, 1.5 M in H2O, Potassium sodium tartrate tetrahydrate, ≧99.5% (NT), Potassium tetraborate tetrahydrate, ≧99.0% (T), Potassium tetraoxalate dihydrate, ≧99.5% (RT), Propionic acid solution, 1.0 M in H2O, STE buffer solution, for molecular biology, pH 7.8, STET buffer solution, for molecular biology, pH 8.0, Sodium 5,5-diethylbarbiturate, ≧99.5% (NT), Sodium acetate solution, for molecular biology, ˜3 M in H2O, Sodium acetate trihydrate, ≧99.5% (NT), Sodium acetate, anhydrous, ≧99.0% (NT), Sodium acetate, for luminescence, anhydrous, ≧99.0% (NT), Sodium acetate, for molecular biology, anhydrous, ≧99.0% (NT), Sodium bicarbonate, ≧99.5% (T), Sodium bitartrate monohydrate, ≧99.0% (T), Sodium carbonate decahydrate, ≧99.5% (T), Sodium carbonate, anhydrous, ≧99.5% (calc. on dry substance, T), Sodium citrate monobasic, anhydrous, ≧99.5% (T), Sodium citrate tribasic dihydrate, ≧99.0% (NT), Sodium citrate tribasic dihydrate, for luminescence, ≧99.0% (NT), Sodium citrate tribasic dihydrate, for molecular biology, ≧99.5% (NT), Sodium formate solution, 8 M in H2O, Sodium oxalate, ≧99.5% (RT), Sodium phosphate dibasic dihydrate, ≧99.0% (T), Sodium phosphate dibasic dihydrate, for luminescence, ≧99.0% (T), Sodium phosphate dibasic dihydrate, for molecular biology, ≧99.0% (T), Sodium phosphate dibasic dodecahydrate, ≧99.0% (T), Sodium phosphate dibasic solution, 0.5 M in H2O, Sodium phosphate dibasic, anhydrous, ≧99.5% (T), Sodium phosphate dibasic, for molecular biology, ≧99.5% (T), Sodium phosphate monobasic dihydrate, ≧99.0% (T), Sodium phosphate monobasic dihydrate, for molecular biology, ≧99.0% (T), Sodium phosphate monobasic monohydrate, for molecular biology, ≧99.5% (T), Sodium phosphate monobasic solution, 5 M in H2O, Sodium pyrophosphate dibasic, ≧99.0% (T), Sodium pyrophosphate tetrabasic decahydrate, ≧99.5% (T), Sodium tartrate dibasic dihydrate, ≧99.0% (NT), Sodium tartrate dibasic solution, 1.5 M in H2O (colorless solution at 20° C.), Sodium tetraborate decahydrate, ≧99.5% (T), TAPS, ≧99.5% (T), TES, ≧99.5% (calc. based on dry substance, T), TM buffer solution, for molecular biology, pH 7.4, TNT buffer solution, for molecular biology, pH 8.0, TRIS Glycine buffer solution, 10× concentrate, TRIS acetate—EDTA buffer solution, for molecular biology, TRIS buffered saline, 10× concentrate, TRIS glycine SDS buffer solution, for electrophoresis, 10× concentrate, TRIS phosphate—EDTA buffer solution, for molecular biology, concentrate, 10× concentrate, Tricine, ≧99.5% (NT), Triethanolamine, ≧99.5% (GC), Triethylamine, ≧99.5% (GC), Triethylammonium acetate buffer, volatile buffer, ˜1.0 M in H2O, Triethylammonium phosphate solution, volatile buffer, ˜1.0 M in H2O, Trimethylammonium acetate solution, volatile buffer, ˜1.0 M in H2O, Trimethylammonium phosphate solution, volatile buffer, ˜1 M in H2O, Tris-EDTA buffer solution, for molecular biology, concentrate, 100× concentrate, Tris-EDTA buffer solution, for molecular biology, pH 7.4, Tris-EDTA buffer solution, for molecular biology, pH 8.0, Trizma® acetate, ≧99.0% (NT), Trizma® base, ≧99.8% (T), Trizma® base, ≧99.8% (T), Trizma® base, for luminescence, ≧99.8% (T), Trizma® base, for molecular biology, ≧99.8% (T), Trizma® carbonate, ≧98.5% (T), Trizma® hydrochloride buffer solution, for molecular biology, pH 7.2, Trizma® hydrochloride buffer solution, for molecular biology, pH 7.4, Trizma® hydrochloride buffer solution, for molecular biology, pH 7.6, Trizma® hydrochloride buffer solution, for molecular biology, pH 8.0, Trizma® hydrochloride, ≧99.0% (AT), Trizma® hydrochloride, for luminescence, ≧99.0% (AT), Trizma® hydrochloride, for molecular biology, ≧99.0% (AT), and Trizma® maleate, ≧99.5% (NT).
  • The nanoemulsion can comprise one or more emulsifying agents to aid in the formation of emulsions. Emulsifying agents include compounds that aggregate at the oil/water interface to form a kind of continuous membrane that prevents direct contact between two adjacent droplets. Certain embodiments of the present invention feature nanoemulsions that may readily be diluted with water to a desired concentration without impairing their antiviral properties.
  • 6. Active Agents Incorporated into a Nanoemulsion of the Invention
  • In a further embodiment of the invention, a nanoemulsion comprises an additional active agent, such as an antibiotic or a palliative agent (such as for burn wound treatment). Addition of another agent may enhance the therapeutic effectiveness of the nanoemulsion. The nanoemulsion in and of itself has anti-bacterial activity and does not need to be combined with another active agent to obtain therapeutic effectiveness. Any antibacterial (or antibiotic) agent suitable for treating a bacterial infection can be incorporated into the topical nanoemulsions of the invention.
  • Examples of such antibiotic agents include, but are not limited to, aminoglycosides, Ansamycins, Carbacephems, Carbapenems, Cephalosporins, Glycopeptides, Macrolides, Monobactams, Penicillins, Polypeptides, Polymyxin, Quinolones, Sulfonamides, Tetracyclines, and others (e.g., Arsphenamine, Chloramphenicol, Clindamycin, Lincomycin, Ethambutol, Fosfomycin, Fusidic acid, Furazolidone, Isoniazid, Linezolid, Metronidazole, Mupirocin, Nitrofurantoin, Platensimycin, Pyrazinamide, Quinupristin/Dalfopristin, Rifampicin (Rifampin in US), Thiamphenicol, Timidazole, Dapsone, and lofazimine).
  • Examples of these classes of antibiotics include, but are not limited to, Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Streptomycin, Tobramycin, Paromomycin, Geldanamycin, Herbimycin, Loracarbef, Ertapenem, Doripenem, Imipenem/Cilastatin, Meropenem, Cefadroxil, Cefazolin, Cefalotin or Cefalothin, Cefalexin, Cefaclor, Cefamandole, Cefoxitin, Cefprozil, Cefuroxime, Cefixime, Cefdinir, Cefditoren, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefepime, Ceftobiprole, Teicoplanin, Vancomycin, Azithromycin, Clarithromycin, Dirithromycin, Erythromycin, Roxithromycin, Troleandomycin, Telithromycin, Spectinomycin, Aztreonam, Amoxicillin, Ampicillin, Azlocillin, Carbenicillin, Cloxacillin, Dicloxacillin, Flucloxacillin, Mezlocillin, Meticillin, Nafcillin, Oxacillin, Penicillin, Piperacillin, Ticarcillin, Bacitracin, Colistin, Polymyxin B, Ciprofloxacin, Enoxacin, Gatifloxacin, Levofloxacin, Lomefloxacin, Moxifloxacin, Norfloxacin, Ofloxacin, Trovafloxacin, Grepafloxacin, Sparfloxacin, Temafloxacin, Mafenide, Sulfonamidochrysoidine (archaic), Sulfacetamide, Sulfadiazine, Sulfamethizole, Sulfanilimide (archaic), Sulfasalazine, Sulfisoxazole, Trimethoprim, rimethoprim-Sulfamethoxazole (Co-trimoxazole) (TMP-SMX), Demeclocycline, Doxycycline, Minocycline, Oxytetracycline, and Tetracycline.
  • Examples of palliative agents which may be incorporated into the nanoemulsions of the invention include, but are not limited to, menthol, camphor, phenol, allantoin, benzocaine, corticosteroids, phenol, zinc oxide, camphor, pramoxine, dimethicone, meradimate, octinoxate, octisalate, oxybenzone, dyclonine, alcohols (e.g., benzyl alcohol), mineral oil, propylene glycol, titanium dioxide, silver nitrate (AgNO3), silver sulfadiazine, mafenide acetate, nanocrystalline impregnated silver dressings, a p38 MAPK inhibitor, and magnesium stearate.
  • D. Pharmaceutical Compositions
  • The nanoemulsions of the invention may be formulated into pharmaceutical compositions that comprise the nanoemulsion in a therapeutically effective amount and suitable, pharmaceutically-acceptable excipients for administration to a human subject in need thereof using any conventional pharmaceutical method of administration. Such excipients are well known in the art.
  • By the phrase “therapeutically effective amount” it is meant any amount of the nanoemulsion that is effective in treating microorganisms by killing or inhibiting the growth of the microorganisms, causing the microorganisms to lose pathogenicity, or any combination thereof.
  • Exemplary dosage forms may include, but are not limited to, patches, ointments, creams, emulsions, liquids, lotions, gels, bioadhesive gels, aerosols, pastes, foams, sunscreens, capsules, microcapsules, or in the form of an article or carrier, such as a bandage, insert, syringe-like applicator, pessary, powder, and talc or other solid.
  • The pharmaceutical compositions may be formulated for immediate release, sustained release, controlled release, delayed release, or any combinations thereof. In some embodiments, the formulations may comprise a penetration-enhancing agent for enhancing penetration of the nanoemulsion through the stratum corneum and into the epidermis or dermis (i.e., for methods of treating burn wounds). Suitable penetration-enhancing agents include, but are not limited to, alcohols such as ethanol, triglycerides and aloe compositions. The amount of the penetration-enhancing agent may comprise from about 0.5% to about 40% by weight of the formulation.
  • When appropriate, for example when treating burn wounds, the nanoemulsions of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis. Such transdermal methods, which comprise applying an electrical current, are well known in the art.
  • In another embodiment of the invention, minimal systemic absorption of the nanoemulsion occurs upon topical administration. Such minimal systemic exposure can be determined by the detection of less than 10 ng/mL, less than 8 ng/mL, less than 5 ng/mL, less than 4 ng/mL, less than 3 ng/mL, or less than 2 ng/mL of the one or more surfactants present in the nanoemulsion in the plasma of the subject. Lack of systemic absorption may be monitored, for example, by measuring the amount of the surfactant, such as the cationic surfactant, in the plasma of the human subject undergoing treatment. Amounts of surfactant of equal to or less than about 10 ng/ml in the plasma confirms minimal systemic absorption.
  • The pharmaceutical compositions may be applied in a single administration or in multiple administrations. The pharmaceutical compositions can be applied for at least one day, at least two days at least three days at least four days at least 5 days, once a week, at least twice a week, at least once a day, at least twice a day, multiple times daily, multiple times weekly, biweekly, at least once a month, or any combination thereof.
  • Following topical or intradermal administration, the nanoemulsion may be occluded or semi-occluded. Occlusion or semi-occlusion may be performed by overlaying a bandage, polyolefin film, article of clothing, impermeable barrier, or semi-impermeable barrier to the topical preparation.
  • Exemplary Nanoemulsions
  • Several exemplary nanoemulsions are described below, although the methods of the invention are not limited to the use of such nanoemulsions. The components and quantity of each can be varied as described herein in the preparation of other nanoemulsions. (“CPC” refers to cetylpyridinium chloride, which is a cationic surfactant present in the nanoemulsions). Compositions are w/w % unless otherwise noted.
  • TABLE 1
    Exemplary Nanoemulsions
    Nanoemulsion Component Weight Percent
    W205EC ED Distilled Water 23.418% 
    EDTA 0.0745% 
    Cetylpyridinium Chloride 1.068%
    Tween
    20  5.92%
    Ethanol  6.73%
    Soybean Oil 62.79%
    P4075EC Distilled Water 23.49%
    CPC 1.068%
    Poloxamer
    407  5.92%
    Ethanol  6.73%
    Soybean Oil, NP 62.79%
    W205GBA2 (v/v %) Distilled Water 20.93%
    BTC 824    2%
    Tween
    20    5%
    Glycerine
       8%
    Soybean Oil   64%
    W805EC Water 23.490% 
    Ethanol 6.730%
    Cetylpyridinium Chloride 1.068%
    Polysorbate
    80 5.920%
    Refined Soybean Oil 62.790% 
    W205ECEDL2 Distilled Water 23.418% 
    EDTA 0.0745% 
    Cetylpyridinium Chloride 1.068%
    Tween
    20  5.92%
    Ethanol  6.73%
    Soybean Oil 62.79%
    W205GBA2ED (v/v %) Distilled Water 20.93%
    EDTA 0.0745% 
    BTC 824    2%
    Tween
    20    5%
    Glycerine
       8%
    Soybean Oil   64%
  • The following nanoemulsions have an average particle (droplet) size of about 300 nm to about 600 nm: W205EC ED, P4075EC, W205GBA2, W805EC, and W205GBA2ED. The W205ECEDL2, which undergoes high pressure processing has an average particle (droplet) size of about 150 nm to about 400 nm. The formulations listed in the table above are “neat” or “concentrated” formulations, meaning that the formulation intended for therapeutic use can be diluted as desired.
  • Methods of Manufacture
  • The nanoemulsions of the invention can be formed using classic emulsion forming techniques. See e.g., U.S. 2004/0043041. See also U.S. Pat. Nos. 6,015,832, 6,506,803, 6,559,189, 6,635,676, and US Patent Publication No. 20040043041, all of which are incorporated by reference. In addition, methods of making emulsions are described in U.S. Pat. Nos. 5,103,497 and 4,895,452 (herein incorporated by reference). In an exemplary method, the oil is mixed with the aqueous phase under relatively high shear forces (e.g., using high hydraulic and mechanical forces) to obtain a nanoemulsion comprising oil droplets having an average diameter of less than about 1000 nm. Some embodiments of the invention employ a nanoemulsion having an oil phase comprising an alcohol such as ethanol. The oil and aqueous phases can be blended using any apparatus capable of producing shear forces sufficient to form an emulsion, such as French Presses or high shear mixers (e.g., FDA approved high shear mixers are available, for example, from Admix, Inc., Manchester, N.H.). Methods of producing such emulsions are described in U.S. Pat. Nos. 5,103,497 and 4,895,452, herein incorporated by reference in their entireties.
  • In an exemplary embodiment, the nanoemulsions used in the methods of the invention comprise droplets of an oily discontinuous phase dispersed in an aqueous continuous phase, such as water. The nanoemulsions of the invention are stable, and do not decompose even after long storage periods. Certain nanoemulsions of the invention are non-toxic and safe when swallowed, inhaled, or contacted to the skin of a subject.
  • The compositions of the invention can be produced in large quantities and are stable for many months at a broad range of temperatures. The nanoemulsion can have textures ranging from that of a semi-solid cream to that of a thin lotion, and can be applied topically by hand, and can be sprayed onto a surface or nebulized.
  • As stated above, at least a portion of the emulsion may be in the form of lipid structures including, but not limited to, unilamellar, multilamellar, and paucliamellar lipid vesicles, micelles, and lamellar phases.
  • The present invention contemplates that many variations of the described nanoemulsions will be useful in the methods of the present invention. To determine if a candidate nanoemulsion is suitable for use with the present invention, three criteria are analyzed. Using the methods and standards described herein, candidate emulsions can be easily tested to determine if they are suitable. First, the desired ingredients are prepared using the methods described herein, to determine if a nanoemulsion can be formed. If a nanoemulsion cannot be formed, the candidate is rejected. Second, the candidate nanoemulsion should form a stable emulsion. A nanoemulsion is stable if it remains in emulsion form for a sufficient period to allow its intended use. For example, for nanoemulsions that are to be stored, shipped, etc., it may be desired that the nanoemulsion remain in emulsion form for months to years. Typical nanoemulsions that are relatively unstable, will lose their form within a day. Third, the candidate nanoemulsion should have efficacy for its intended use. For example, the emulsions of the invention should kill or disable microorganisms in vitro. To determine the suitability of a particular candidate nanoemulsion against a desired microorganism, the nanoemulsion is exposed to the microorganism for one or more time periods in a side-by-side experiment with an appropriate control sample (e.g., a negative control such as water) and determining if, and to what degree, the nanoemulsion kills or disables the microorganism.
  • The nanoemulsion of the invention can be provided in many different types of containers and delivery systems. For example, in some embodiments of the invention, the nanoemulsions are provided as a liquid, lotion, cream or other solid or semi-solid form. The nanoemulsions of the invention may be incorporated into hydrogel formulations.
  • The nanoemulsions can be delivered (e.g., to a subject or customers) in any suitable container. Suitable containers can be used that provide one or more single use or multi-use dosages of the nanoemulsion for the desired application. In some embodiments of the invention, the nanoemulsions are provided in a suspension or liquid form. Such nanoemulsions can be delivered in any suitable container including spray bottles (e.g., pressurized spray bottles, nebulizers).
  • Exemplary Methods of Use
  • As described in more detail throughout this application, the present invention is directed to methods of treating and/or preventing a respiratory infection in a subject having Cystic fibrosis (CF). In general, the method comprises administering a nanoemulsion to the subject, wherein the nanoemulsion comprises: (i) water; (ii) at least one organic solvent; (iii) at least one surfactant; and (iv) at least one oil; and wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm. In one embodiment of the invention, the subject is susceptible to or has an infection by one or more bacterial species selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp. The nanoemulsion can be delivered using any pharmaceutically acceptable means, with inhalation being one example of a useful administration method.
  • In yet another embodiment, the invention is directed to a method of treating or preventing an infection in a subject having a burn wound, wherein: (a) the method comprises administering a nanoemulsion to the subject; and (b) the nanoemulsion comprises: (i) water; (ii) at least one organic solvent; (iii) at least one surfactant; and (iv) at least one oil; and wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm. In one embodiment of the invention, the subject is susceptible to or has an infection by one or more gram-negative or gram-positive bacterial species. In another embodiment, the bacterial species are selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp. The nanoemulsion can be delivered using any pharmaceutically acceptable means, with inhalation, nebulization, and topical application to mucosal surfaces being examples of useful administration methods.
  • In yet another embodiment, the invention is directed to a method of treating or preventing an Haemophilus influenzae infection in a subject wherein: (a) the method comprises administering a nanoemulsion to the subject having or at risk of having a Haemophilus influenzae infection; (b) the nanoemulsion comprises: (i) water; (ii) at least one organic solvent; (iii) at least one surfactant; and (iv) at least one oil; and (c) wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm. The nanoemulsion can be delivered using any pharmaceutically acceptable means, with inhalation, nebulization, and delivery to a mucosal surface being examples of useful administration methods.
  • In one embodiment of the invention, the nanoemulsion exhibits minimal or no toxicity or side effects. Preferably, the nanoemulsion does not exhibit resistance to bacteria. This embodiment applies to all methods described herein.
  • If the method relates to a respiratory infection, then in one embodiment the respiratory infection may be associated with a bacterial biofilm, such as a biofilm present in the lungs of a subject.
  • All of the methods of the invention may further comprise administering one or more antibiotics either before, during, or after administration of the nanoemulsion. In yet another embodiment, one or more antibiotics may be incorporated into a nanoemulsion. In yet another embodiment of the invention, the nanoemulsion does not exhibit any antagonism with the antibiotic.
  • In one embodiment of the invention, administration of a nanoemulsion and at least one antibiotic is synergistic as defined by a fractional inhibitory concentration (FIC) index, a fractional bactericidal concentration (FBC) index, or a combination thereof. This embodiment applies to all methods described herein. Examples of such antibiotics include, but are not limited to polymyxins (colistin) and aminoglycosides (tobramycin).
  • In yet another embodiment, the methods of the invention may be used to treat or prevent infection by one or more bacterial species selected from the group consisting of Pseudomonas aeruginosa, B. cenocepacia, A. baumannii, Stenotrophomonas maltophilia, Staphylococcus aureus, H influenzae, E. coli, K. pneumoniae, and Proteus mirabilis. All other gram positive or gram negative bacteria are also encompassed by the methods of the invention.
  • In one embodiment, the minimum inhibitory concentration (MIC), the minimum bactericidal concentration (MBC), or a combination thereof for the nanoemulsion demonstrate bacteriostatic or bactericidal activity for the nanoemulsion. This embodiment applies to all methods described herein.
  • In another embodiment of the invention, one or more bacterial species may exhibit resistance against one or more antibiotics. For example, the bacterial species can be methicillin-resistant Staphylococcus aureus (MRSA). This embodiment applies to all methods described herein.
  • The present invention is not limited by the type of subject administered a composition of the present invention. Each of the subjects (e.g., susceptible to respiratory infection) described above may be administered a composition of the present invention. In addition, the compositions and methods of the present invention are useful in the treatment of other respiratory diseases and disorders, such as acute bronchitis, bronchiectasis, pneumonia (including ventilator-associated pneumonia, nosocomial pneumonia, viral pneumonia, bacterial pneumonia, mycobacterial pneumonia, fungal pneumonia, eosinophilic pneumonia, and Pneumocystis carinii pneumonia), tuberculosis, cystic fibrosis (CF), emphysema radiation pneumonitis, and respiratory infection associated with inflammation caused by smoking, pulmonary edema, pneumoconiosis, sarcoidiosis, silicosis, asbestosis, berylliosis, coal worker's pneumonoconiosis (CWP), byssinosis, interstitial lung diseases (ILD) such as idiopathic pulmonary fibrosis, ILD associated with collagen vascular disorders, systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, systemic sclerosis, and pulmonary inflammation that is a result of or is secondary to another disorder such as influenza.
  • The present invention is not limited by the particular formulation of a composition comprising a nanoemulsion of the present invention. Indeed, a composition comprising a nanoemulsion of the present invention may comprise one or more different agents in addition to the nanoemulsion. These agents or cofactors include, but are not limited to, adjuvants, surfactants, additives, buffers, solubilizers, chelators, oils, salts, therapeutic agents, drugs, bioactive agents, antibacterials, and antimicrobial agents (e.g., antibiotics, antivirals, etc.). In some embodiments, a composition comprising a nanoemulsion of the present invention comprises an agent and/or co-factor that enhance the ability of the nanoemulsion to kill a microbe (e.g., located in the respiratory tract). In some preferred embodiments, the presence of one or more co-factors or agents reduces the amount of nanoemulsion required for killing and/or attenuation of growth of a microbe. The present invention is not limited by the type of co-factor or agent used in a therapeutic agent of the present invention.
  • In some embodiments, a co-factor or agent used in a nanoemulsion composition is a bioactive agent. For example, in some embodiments, the bioactive agent may be a bioactive agent useful in a cell (e.g., a cell expressing a CFTR). Bioactive agents, as used herein, include diagnostic agents such as radioactive labels and fluorescent labels. Bioactive agents also include molecules affecting the metabolism of a cell (e.g., a cell expressing a CFTR), including peptides, nucleic acids, and other natural and synthetic drug molecules. Bioactive agents include, but are not limited to, adrenergic agent; adrenocortical steroid; adrenocortical suppressant; alcohol deterrent; aldosterone antagonist; amino acid; ammonia detoxicant; anabolic; analeptic; analgesic; androgen; anesthesia, adjunct to; anesthetic; anorectic; antagonist; anterior pituitary suppressant; anthelmintic; anti-acne agent; anti-adrenergic; anti-allergic; anti-amebic; anti-androgen; anti-anemic; anti-anginal; anti-anxiety; anti-arthritic; anti-asthmatic; anti-atherosclerotic; antibacterial; anticholelithic; anticholelithogenic; anticholinergic; anticoagulant; anticoccidal; anticonvulsant; antidepressant; antidiabetic; antidiarrheal; antidiuretic; antidote; anti-emetic; anti-epileptic; anti-estrogen; antifibrinolytic; antifungal; antiglaucoma agent; antihemophilic; antihemorrhagic; antihistamine; antihyperlipidemia; antihyperlipoproteinemic; antihypertensive; antihypotensive; anti-infective; anti-infective, topical; anti-inflammatory; antikeratinizing agent; antimalarial; antimicrobial; antimigraine; antimitotic; antimycotic, antinauseant, antineoplastic, antineutropenic, antiobessional agent; antiparasitic; antiparkinsonian; antiperistaltic, antipneumocystic; antiproliferative; antiprostatic hypertrophy; antiprotozoal; antipruritic; antipsychotic; antirheumatic; antischistosomal; antiseborrheic; antisecretory; antispasmodic; antithrombotic; antitussive; anti-ulcerative; anti-urolithic; antiviral; appetite suppressant; benign prostatic hyperplasia therapy agent; blood glucose regulator; bone resorption inhibitor; bronchodilator; carbonic anhydrase inhibitor; cardiac depressant; cardioprotectant; cardiotonic; cardiovascular agent; choleretic; cholinergic; cholinergic agonist; cholinesterase deactivator; coccidiostat; cognition adjuvant; cognition enhancer; depressant; diagnostic aid; diuretic; dopaminergic agent; ectoparasiticide; emetic; enzyme inhibitor; estrogen; fibrinolytic; fluorescent agent; free oxygen radical scavenger; gastrointestinal motility effector; glucocorticoid; gonad-stimulating principle; hair growth stimulant; hemostatic; histamine H2 receptor antagonists; hormone; hypocholesterolemic; hypoglycemic; hypolipidemic; hypotensive; imaging agent; immunizing agent; immunomodulator; immunoregulator; immunostimulant; immunosuppressant; impotence therapy adjunct; inhibitor; keratolytic; LHRH agonist; liver disorder treatment; luteolysin; memory adjuvant; mental performance enhancer; mood regulator; mucolytic; mucosal protective agent; mydriatic; nasal decongestant; neuromuscular blocking agent; neuroprotective; NMDA antagonist; non-hormonal sterol derivative; oxytocic; plasminogen activator; platelet activating factor antagonist; platelet aggregation inhibitor; post-stroke and post-head trauma treatment; potentiator; progestin; prostaglandin; prostate growth inhibitor; prothyrotropin; psychotropic; pulmonary surface; radioactive agent; regulator; relaxant; repartitioning agent; scabicide; sclerosing agent; sedative; sedative-hypnotic; selective adenosine A1 antagonist; serotonin antagonist; serotonin inhibitor; serotonin receptor antagonist; steroid; stimulant; suppressant; symptomatic multiple sclerosis; synergist; thyroid hormone; thyroid inhibitor; thyromimetic; tranquilizer; amyotrophic lateral sclerosis agent; cerebral ischemia agent; Paget's disease agent; unstable angina agent; uricosuric; vasoconstrictor; vasodilator; vulnerary; wound healing agent; xanthine oxidase inhibitor.
  • Molecules useful as antimicrobials can be delivered by the methods and compositions of the invention, such that the respiratory infection is reduced or eliminated. Antibiotics that may find use in co-administration with a composition comprising a nanoemulsion of the present invention include, but are not limited to, agents or drugs that are bactericidal and/or bacteriostatic (e.g., inhibiting replication of bacteria or inhibiting synthesis of bacterial components required for survival of the infecting organism), including, but not limited to, almecillin, amdinocillin, amikacin, amoxicillin, amphomycin, amphotericin B, ampicillin, azacitidine, azaserine, azithromycin, azlocillin, aztreonam, bacampicillin, bacitracin, benzyl penicilloyl-polylysine, bleomycin, candicidin, capreomycin, carbenicillin, cefaclor, cefadroxil, cefamandole, cefazoline, cefdinir, cefepime, cefixime, cefinenoxime, cefinetazole, cefodizime, cefonicid, cefoperazone, ceforanide, cefotaxime, cefotetan, cefotiam, cefoxitin, cefpiramide, cefpodoxime, cefprozil, cefsulodin, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefuroxime, cephacetrile, cephalexin, cephaloglycin, cephaloridine, cephalothin, cephapirin, cephradine, chloramphenicol, chlortetracycline, cilastatin, cinnamycin, ciprofloxacin, clarithromycin, clavulanic acid, clindamycin, clioquinol, cloxacillin, colistimethate, colistin, cyclacillin, cycloserine, cyclosporine, cyclo-(Leu-Pro), dactinomycin, dalbavancin, dalfopristin, daptomycin, daunorubicin, demeclocycline, detorubicin, dicloxacillin, dihydrostreptomycin, dirithromycin, doxorubicin, doxycycline, epirubicin, erythromycin, eveminomycin, floxacillin, fosfomycin, fusidic acid, gemifloxacin, gentamycin, gramicidin, griseofulvin, hetacillin, idarubicin, imipenem, iseganan, ivermectin, kanamycin, laspartomycin, linezolid, linocomycin, loracarbef, magainin, meclocycline, meropenem, methacycline, methicillin, mezlocillin, minocycline, mitomycin, moenomycin, moxalactam, moxifloxacin, mycophenolic acid, nafcillin, natamycin, neomycin, netilmicin, niphimycin, nitrofurantoin, novobiocin, oleandomycin, oritavancin, oxacillin, oxytetracycline, paromomycin, penicillamine, penicillin G, penicillin V, phenethicillin, piperacillin, plicamycin, polymyxin B, pristinamycin, quinupristin, rifabutin, rifampin, rifamycin, rolitetracycline, sisomicin, spectrinomycin, streptomycin, streptozocin, sulbactam, sultamicillin, tacrolimus, tazobactam, teicoplanin, telithromycin, tetracycline, ticarcillin, tigecycline, tobramycin, troleandomycin, tunicamycin, tyrthricin, vancomycin, vidarabine, viomycin, virginiamycin, BMS-284,756, L-749,345, ER-35,786, S-4661, L-786,392, MC-02479, Pep5, RP 59500, and TD-6424.
  • In some embodiments, a composition comprising a nanoemulsion of the present invention comprises one or more mucoadhesives (See, e.g., U.S. Pat. App. No. 20050281843, hereby incorporated by reference in its entirety). The present invention is not limited by the type of mucoadhesive utilized. Indeed, a variety of mucoadhesives are contemplated to be useful in the present invention including, but not limited to, cross-linked derivatives of poly(acrylic acid) (e.g., carbopol and polycarbophil), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides (e.g., alginate and chitosan), hydroxypropyl methylcellulose, lectins, fimbrial proteins, and carboxymethylcellulose. Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, use of a mucoadhesive (e.g., in a composition comprising a nanoemulsion) enhances killing and/or attenuation of growth of a microbe (e.g., exposed to a composition of the present invention) due to an increase in duration and/or amount of exposure to the nanoemulsion that a subject and/or microbe experiences when a mucoadhesive is used compared to the duration and/or amount of exposure to the nanoemulsion in the absence of using the mucoadhesive.
  • In some embodiments, a composition of the present invention may comprise sterile aqueous preparations. Acceptable vehicles and solvents include, but are not limited to, water, Ringer's solution, phosphate buffered saline and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed mineral or non-mineral oil may be employed including synthetic mono-ordi-glycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Carrier formulations suitable for mucosal, pulmonary, subcutaneous, intramuscular, intraperitoneal, intravenous, or administration via other routes may be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • A composition comprising a nanoemulsion of the present invention can be used therapeutically (e.g., to kill and/or attenuate growth of an existing infection) or as a prophylactic (e.g., to prevent microbial growth and/or colonization (e.g., to prevent signs or symptoms of disease)). A composition comprising a nanoemulsion of the present invention can be administered to a subject via a number of different delivery routes and methods.
  • For example, the compositions of the present invention can be administered to a subject (e.g., mucosally or by pulmonary route) by multiple methods, including, but not limited to: being suspended in a solution and applied to a surface; being suspended in a solution and sprayed onto a surface using a spray applicator; being mixed with a mucoadhesive and applied (e.g., sprayed or wiped) onto a surface (e.g., mucosal or pulmonary surface); being placed on or impregnated onto a nasal and/or pulmonary applicator and applied; being applied by a controlled-release mechanism; applied using a nebulizer, aerosolized, being applied as a liposome; or being applied on a polymer.
  • In some embodiments, compositions of the present invention are administered mucosally (e.g., using standard techniques; See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995 (e.g., for mucosal delivery techniques, including intranasal and pulmonary techniques), as well as European Publication No. 517,565 and Illum et al., J. Controlled Rel., 1994, 29:133-141 (e.g., for techniques of intranasal administration), each of which is hereby incorporated by reference in its entirety). The present invention is not limited by the route of administration.
  • Methods of intranasal and pulmonary administration are well known in the art, including the administration of a droplet or spray form of the nanoemulsion into the nasopharynx of a subject to be treated. In some embodiments, a nebulized or aerosolized composition comprising a nanoemulsion is provided. Enteric formulations such as gastro resistant capsules for oral administration, suppositories for rectal or vaginal administration may also form part of this invention. Compositions of the present invention may also be administered via the oral route. Under these circumstances, a composition comprising a nanoemulsion may comprise a pharmaceutically acceptable excipient and/or include alkaline buffers, or enteric capsules. Formulations for nasal delivery may include those with dextran or cyclodextran and saponin as an adjuvant.
  • In preferred embodiments, a nanoemulsion of the present invention is administered via a pulmonary delivery route and/or means. In some embodiments, an aqueous solution containing the nanoemulsion is gently and thoroughly mixed to form a solution. The solution is sterile filtered (e.g., through a 0.2 micron filter) into a sterile, enclosed vessel. Under sterile conditions, the solution is passed through an appropriately small orifice to make droplets (e.g., between 0.1 and 10 microns).
  • The particles may be administered using any of a number of different applicators. Suitable methods for manufacture and administration are described in the following U.S. Pat. Nos. 6,592,904; 6,518,239; 6,423,344; 6,294,204; 6,051,256 and 5,997,848 to INHALE (now NEKTAR); and U.S. Pat. No. 5,985,309; RE37,053; U.S. Pat. Nos. 6,436,443; 6,447,753; 6,503,480; and 6,635,283, to Edwards, et al. (MIT, AIR), each of which is hereby incorporated
  • Thus, in some embodiments, compositions of the present invention are administered by pulmonary delivery. For example, a composition of the present invention can be delivered to the lungs of a subject (e.g., a human) via inhalation (See, e.g., Adjei, et al. Pharmaceutical Research 1990; 7:565-569; Adjei, et al. Int. J. Pharmaceutics 1990; 63:135-144; Braquet, et al. J. Cardiovascular Pharmacology 1989 143-146; Hubbard, et al. (1989) Annals of Internal Medicine, Vol. III, pp. 206-212; Smith, et al. J. Clin. Invest. 1989; 84:1145-1146; Oswein, et al. “Aerosolization of Proteins”, 1990; Proceedings of Symposium on Respiratory Drug Delivery II Keystone, Colo.; Debs, et al. J. Immunol. 1988; 140:3482-3488; and U.S. Pat. No. 5,284,656 to Platz, et al, each of which are hereby incorporated by reference in its entirety). A method and composition for pulmonary delivery of drugs for systemic effect is described in U.S. Pat. No. 5,451,569 to Wong, et al., hereby incorporated by reference; See also U.S. Pat. No. 6,651,655 to Licalsi et al., hereby incorporated by reference in its entirety)). In some embodiments, a composition comprising a nanoemulsion is administered to a subject by more than one route or means (e.g., administered via pulmonary route as well as a mucosal route).
  • Further contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary and/or nasal mucosal delivery of pharmaceutical agents including, but not limited to, nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art. Some specific examples of commercially available devices suitable for the practice of this invention are the ULTRAVENT nebulizer (Mallinckrodt Inc., St. Louis, Mo.); the ACORN II nebulizer (Marquest Medical Products, Englewood, Colo.); the VENTOLIN metered dose inhaler (Glaxo Inc., Research Triangle Park, N.C.); and the SPINHALER powder inhaler (Fisons Corp., Bedford, Mass.). All such devices require the use of formulations suitable for dispensing of the therapeutic agent. Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants, surfactants, carriers and/or other agents useful in therapy. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated.
  • Thus, in some embodiments, a composition comprising a nanoemulsion of the present invention may be used to protect and/or treat a subject susceptible to, or suffering from, a disease by means of administering compositions comprising a nanoemulsion by mucosal, intramuscular, intraperitoneal, intradermal, transdermal, pulmonary, intravenous, subcutaneous or other route of administration described herein. Methods of systemic administration of the nanoemulsion and/or agent co-administered with the nanoemulsion may include conventional syringes and needles, or devices designed for ballistic delivery (See, e.g., WO 99/27961, hereby incorporated by reference), or needleless pressure liquid jet device (See, e.g., U.S. Pat. No. 4,596,556; U.S. Pat. No. 5,993,412, each of which are hereby incorporated by reference), or transdermal patches (See, e.g., WO 97/48440; WO 98/28037, each of which are hereby incorporated by reference). In some embodiments, the present invention provides a delivery device for systemic administration, pre-filled with the nanoemulsion composition of the present invention.
  • As described above, the present invention is not limited by the type of subject administered a composition of the present invention. Indeed, a wide variety of subjects are contemplated to be benefited from administration of a composition of the present invention. In preferred embodiments, the subject is a human. In some embodiments, human subjects are of any age (e.g., adults, children, infants, etc.) that have been or are likely to become exposed to a microorganism. In some embodiments, the human subjects are subjects that are more likely to receive a direct exposure to pathogenic microorganisms or that are more likely to display signs and symptoms of disease after exposure to a pathogen (e.g., subjects with CF or asthma, subjects in the armed forces, government employees, frequent travelers, persons attending or working in a school or daycare, health care workers, an elderly person, an immunocompromised person, and emergency service employees (e.g., police, fire, EMT employees)). In some embodiments, any one or all members of the general public can be administered a composition of the present invention (e.g., to prevent the occurrence or spread of disease). For example, in some embodiments, compositions and methods of the present invention are utilized to treat a group of people (e.g., a population of a region, city, state and/or country) for their own health (e.g., to prevent or treat disease) and/or to prevent or reduce the risk of disease spread from animals (e.g., birds, cattle, sheep, pigs, etc.) to humans. In some embodiments, the subjects are non-human mammals (e.g., pigs, cattle, goats, horses, sheep, or other livestock; or mice, rats, rabbits or other animal). In some embodiments, compositions and methods of the present invention are utilized in research settings (e.g., with research animals).
  • A composition comprising a nanoemulsion of the present invention can be administered (e.g., to a subject (e.g., via pulmonary and/or mucosal route) or to microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria (e.g., residing on or within the respiratory system of a subject and/or on or within a burn wound)))) as a therapeutic or as a prophylactic to prevent microbial infection. Thus, in some embodiments, the present invention provides a method of altering microbial (e.g., bacterial (e.g., opportunistic and/or pathogenic bacterial) growth comprising administering a composition comprising a nanoemulsion to the microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria). In some embodiments, administration of a composition comprising a nanoemulsion to the microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) kills the microbes. In some embodiments, administration of a composition comprising nanoemulsion to the microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) inhibits growth of the microbes. It is contemplated that a composition comprising a nanoemulsion can be administered to microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria (e.g., residing within the respiratory tract))) via a number of delivery routes and/or mechanisms.
  • The compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions. Thus, for example, the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, preferably do not unduly interfere with the biological activities of the components of the compositions of the present invention. The formulations can be sterilized and, if desired, mixed with auxiliary agents (e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like) that do not deleteriously interact with the nanoemulsion. In some embodiments, nanoemulsion compositions of the present invention are administered in the form of a pharmaceutically acceptable salt. When used the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include, but are not limited to, acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v). Suitable preservatives may include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • In some embodiments, a composition comprising a nanoemulsion is co-administered with one or more antibiotics. For example, one or more antibiotics may be administered with, before and/or after administration of a composition comprising a nanoemulsion. The present invention is not limited by the type of antibiotic co-administered. Indeed, a variety of antibiotics may be co-administered including, but not limited to, β-lactam antibiotics, penicillins (such as natural penicillins, aminopenicillins, penicillinase-resistant penicillins, carboxy penicillins, ureido penicillins), cephalosporins (first generation, second generation, and third generation cephalosporins), and other β-lactams (such as imipenem, monobactams), β-lactamase inhibitors, vancomycin, aminoglycosides and spectinomycin, tetracyclines, chloramphenicol, erythromycin, lincomycin, clindamycin, rifampin, metronidazole, polymyxins, doxycycline, quinolones (e.g., ciprofloxacin), sulfonamides, trimethoprim, and quinolines.
  • A wide variety of antimicrobial agents are currently available for use in treating bacterial, fungal and viral infections. For a comprehensive treatise on the general classes of such drugs and their mechanisms of action, the skilled artisan is referred to Goodman & Gilman's “The Pharmacological Basis of Therapeutics” Eds. Hardman et al., 9th Edition, Pub. McGraw Hill, chapters 43 through 50, 1996, (herein incorporated by reference in its entirety). Generally, these agents include agents that inhibit cell wall synthesis (e.g., penicillins, cephalosporins, cycloserine, vancomycin, bacitracin); and the imidazole antifungal agents (e.g., miconazole, ketoconazole and clotrimazole); agents that act directly to disrupt the cell membrane of the microorganism (e.g., detergents such as polmyxin and colistimethate and the antifungals nystatin and amphotericin B); agents that affect the ribosomal subunits to inhibit protein synthesis (e.g., chloramphenicol, the tetracyclines, erthromycin and clindamycin); agents that alter protein synthesis and lead to cell death (e.g., aminoglycosides); agents that affect nucleic acid metabolism (e.g., the rifamycins and the quinolones); the antimetabolites (e.g., trimethoprim and sulfonamides); and the nucleic acid analogues such as zidovudine, gangcyclovir, vidarabine, and acyclovir which act to inhibit viral enzymes essential for DNA synthesis. Various combinations of antimicrobials may be employed.
  • The present invention also includes methods involving co-administration of a composition comprising a nanoemulsion with one or more additional active and/or anti-infective agents. In co-administration procedures, the agents may be administered concurrently or sequentially. In one embodiment, the compositions described herein are administered prior to the other active agent(s). The pharmaceutical formulations and modes of administration may be any of those described herein. In addition, the two or more co-administered agents may each be administered using different modes (e.g., routes) or different formulations. The additional agents to be co-administered (e.g., antibiotics, a second type of nanoemulsion, etc.) can be any of the well-known agents in the art, including, but not limited to, those that are currently in clinical use.
  • In some embodiments, a composition comprising a nanoemulsion is administered to a subject via more than one route. For example, a subject may benefit from receiving mucosal administration (e.g., nasal administration or other mucosal routes described herein) and, additionally, receiving one or more other routes of administration (e.g., pulmonary administration (e.g., via a nebulizer, inhaler, or other methods described herein.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the compositions, increasing convenience to the subject and a physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer based systems such as poly (lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.
  • Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109, hereby incorporated by reference. Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like. Specific examples include, but are not limited to: (a) erosional systems in which an agent of the invention is contained in a form within a matrix such as those described in U.S. Pat. Nos. 4,452,775, 4,675,189, and 5,736,152, each of which is hereby incorporated by reference and (b) diffusional systems in which an active component permeates at a controlled rate from a polymer such as described in U.S. Pat. Nos. 3,854,480, 5,133,974 and 5,407,686, each of which is hereby incorporated by reference. In addition, pump-based hardware delivery systems can be used, some of which are adapted for implantation.
  • In preferred embodiments, a composition comprising a nanoemulsion of the present invention comprises a suitable amount of the nanoemulsion to kill and/or attenuate growth of microbes (e.g., pathogenic microbes (e.g., pathogenic bacteria, viruses, etc.)) in a subject when administered to the subject. The present invention is not limited by the amount of nanoemulsion used. In some preferred embodiments, the amount of nanoemulsion in a composition comprising a nanoemulsion is selected as that amount which kills and/or attenuates microbial growth without significant, adverse side effects. The amount will vary depending upon which specific nanoemulsion(s) is/are employed, and can vary from subject to subject, depending on a number of factors including, but not limited to, the species, age and general condition (e.g., health) of the subject, and the mode of administration. Procedures for determining the appropriate amount of nanoemulsion administered to a subject to kill and/or attenuate growth of a microbe (e.g., pathogenic microbe (e.g., pathogenic bacteria, viruses, etc.)) in a subject can be readily determined using known means by one of ordinary skill in the art.
  • In some embodiments, it is expected that each dose (e.g., of a composition comprising a nanoemulsion (e.g., administered to a subject to kill and/or attenuate microbial growth)) comprises 10-40% nanoemulsion, in some embodiments, 20% nanoemulsion, in some embodiments less than 20% (e.g., 15%, 10%, 8%, 5% or less nanoemulsion), and in some embodiments greater than 20% nanoemulsion (e.g., 25%, 30%, 35%, 40% or more nanoemulsion). An optimal amount for a particular administration (e.g., to kill and/or attenuate microbial growth) can be ascertained by one of skill in the art using standard studies involving observation of microbial growth and/or death and other responses in subjects.
  • In some embodiments, it is expected that each dose (e.g., of a composition comprising a nanoemulsion (e.g., administered to a subject to kill and/or attenuate microbial growth)) is from 0.001 to 40% or more (e.g., 0.001-10%, 0.5-5%, 1-3%, 2%, 6%, 10%, 15%, 20%, 30%, 40% or more) by weight nanoemulsion.
  • Similarly, the present invention is not limited by the duration of time a nanoemulsion is administered to a subject (e.g., to kill and/or attenuate microbial growth). In some embodiments, a nanoemulsion is administered one or more times (e.g. twice, three times, four times or more) daily. In some embodiments, a composition comprising a nanoemulsion is administered one or more times a day until an infection is eradicated or microbial growth and/or presence has been reduced to a desired level. In some embodiments, a composition comprising a nanoemulsion of the present invention is formulated in a concentrated dose that can be diluted prior to administration to a subject. For example, dilutions of a concentrated composition may be administered to a subject such that the subject receives any one or more of the specific dosages provided herein. In some embodiments, dilution of a concentrated composition may be made such that a subject is administered (e.g., in a single dose) a composition comprising 0.5-50% of the nanoemulsion present in the concentrated composition. Concentrated compositions are contemplated to be useful in a setting in which large numbers of subjects may be administered a composition of the present invention (e.g., a hospital). In some embodiments, a composition comprising a nanoemulsion of the present invention (e.g., a concentrated composition) is stable at room temperature for more than 1 week, in some embodiments for more than 2 weeks, in some embodiments for more than 3 weeks, in some embodiments for more than 4 weeks, in some embodiments for more than 5 weeks, and in some embodiments for more than 6 weeks.
  • Dosage units may be proportionately increased or decreased based on several factors including, but not limited to, the weight, age, and health status of the subject. In addition, dosage units may be increased or decreased for subsequent administrations.
  • In some embodiments, a composition comprising a nanoemulsion is administered to a subject under conditions such that microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria)) are killed. In some embodiments, a composition comprising a nanoemulsion is administered to a subject under conditions such that microbial (e.g., bacterial (e.g., opportunistic and/or pathogenic bacterial) growth is prohibited and/or attenuated. In some embodiments, greater than 90% (e.g., greater than 95%, 98%, 99%, all detectable) of microbes (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) are killed. In some embodiments, there is greater than 2 log (e.g., greater than 3 log, 4 log, 5 log, or more) reduction in microbe (e.g., bacteria (e.g., opportunistic and/or pathogenic bacteria) presence. In some embodiments, reduction and/or killing is observed in one hour or less (e.g., 45 minutes, 30 minutes, 15 minutes, or less). In some embodiments, reduction and/or killing is observed in 6 hours or less (e.g., 5 hours, 4, hours, 3 hours, two hours or less than one hour). In some embodiments, reduction and/or killing is observed in two days or less following initial treatment (e.g., less than 24 hours, less than 20 hours, 18 hours or less). In some embodiments, the reduction and/or killing is observed in three days or less, four days or less, or five days or less.
  • A composition comprising a nanoemulsion of the present invention finds use where the nature of the infectious and/or disease causing agent (e.g., causing signs, symptoms or indications of respiratory infection) is known, as well as where the nature of the infectious and/or disease causing agent is unknown (e.g., in emerging disease (e.g., of pandemic proportion (e.g., influenza or other outbreaks of disease))). For example, the present invention contemplates use of the compositions of the present invention in treatment of or prevention of infections associated with an emergent infectious and/or disease causing agent yet to be identified (e.g., isolated and/or cultured from a diseased person but without genetic, biochemical or other characterization of the infectious and/or disease causing agent).
  • It is contemplated that the compositions and methods of the present invention will find use in various settings, including research settings. For example, compositions and methods of the present invention also find use in studies of the immune system (e.g., characterization of adaptive immune responses (e.g., protective immune responses (e.g., mucosal or systemic immunity))). Uses of the compositions and methods provided by the present invention encompass human and non-human subjects and samples from those subjects, and also encompass research applications using these subjects. Compositions and methods of the present invention are also useful in studying and optimizing nanoemulsions, immunogens, and other components and for screening for new components. Thus, it is not intended that the present invention be limited to any particular subject and/or application setting.
  • The formulations can be tested in vivo in a number of animal models developed for the study of pulmonary, mucosal and other routes of delivery. As is readily apparent, the compositions of the present invention are useful for preventing and/or treating a wide variety of diseases and infections caused by viruses, bacteria, parasites, and fungi. Not only can the compositions be used prophylactically or therapeutically, as described above, the compositions can also be used in order to prepare antibodies, both polyclonal and monoclonal (e.g., for diagnostic purposes), as well as for immunopurification of an antigen of interest.
  • In some embodiments, the present invention provides a kit comprising a composition comprising a nanoemulsion. In some embodiments, the kit further provides a device for administering the composition. The present invention is not limited by the type of device included in the kit. In some embodiments, the device is configured for pulmonary application of the composition of the present invention (e.g., a nasal inhaler or nasal mister). In some embodiments, a kit comprises a composition comprising a nanoemulsion in a concentrated form (e.g., that can be diluted prior to administration to a subject).
  • In some embodiments, all kit components are present within a single container (e.g., vial or tube). In some embodiments, each kit component is located in a single container (e.g., vial or tube). In some embodiments, one or more kit components are located in a single container (e.g., vial or tube) with other components of the same kit being located in a separate container (e.g., vial or tube). In some embodiments, a kit comprises a buffer. In some embodiments, the kit further comprises instructions for use.
  • Therapeutic and Prophylactic Use of Immunogenic Compositions
  • The present invention provides immunogenic nanoemulsion compositions and methods of using the same for the induction of immune responses (e.g., innate and/or adaptive immune responses (e.g., for generation of host immunity against a bacterial species of the genus Burkholderia (e.g., B. cenocepacia, B. multivorans, etc.))). Compositions and methods of the present invention find use in, among other things, clinical (e.g. therapeutic and preventative medicine (e.g., vaccination)) and research applications.
  • As shown in Example 10, an immunogenic composition (e.g., comprising a nanoemulsion and Bukholderia antigen) of the present invention induces (e.g., when administered to a subject) both systemic and mucosal immunity. Thus, in some preferred embodiments, administration of a composition comprising a nanoemulsion and Burkholderia antigen to a subject results in protection against an exposure (e.g., a mucosal and/or respiratory exposure) to Burkholderia. Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, mucosal administration (e.g., vaccination) provides protection against Burkholderia (e.g., Burkholderia cepacia) infection (e.g., that initiates at a mucosal surface). Although it has heretofore proven difficult to stimulate secretory IgA responses and protection against pathogens that invade at mucosal surfaces (See, e.g., Mestecky et al, Mucosal Immunology. 3ed edn. (Academic Press, San Diego, 2005)), in some embodiments, the present invention provides compositions and methods for stimulating mucosal immunity (e.g., a protective IgA response) towards a pathogen (e.g., pathogenic species of Burkholderia, Haemophilus, Staphylococcus or other bacterial genus) in a subject.
  • In some embodiments, the present invention provides a composition (e.g., a composition comprising a nanoemulsion and Burkholderia (e.g., Burkholderia cenocepacia) antigen (e.g., an immunogenic polypeptide comprising an amino acid sequence of SEQ ID NO. 1 or SEQ ID NOS. 2-16)) to serve as a mucosal vaccine. In some embodiments, an immunogenic composition comprising nanoemulsion (NE) and Burkholderia antigen is produced with NE and killed whole cell bacteria of the genus Burkholderia (e.g., Burkholderia cepacia (e.g., killed using nanoemulsion, alcohol (e.g., ethanol), or other methods), isolated, purified and/or recombinant protein and/or saccharide component of Burkholderia (e.g., protein/peptide (e.g., Burkholderia-derived protein, live-virus-vector-derived protein, recombinant protein, recombinant denatured protein/antigens, small peptide segments protein/antigen).
  • In some preferred embodiments, the present invention provides a composition for generating an immune response comprising a NE and an immunogen (e.g., a purified, isolated or synthetic Burkholderia protein or derivative, variant, or analogue thereof; or, one or more species of Burkholderia (e.g., Burkholderia multivorans (e.g., killed and or inactivated whole cell bacteria). When administered to a subject, a composition of the present invention stimulates an immune response against the immunogen within the subject. Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, generation of an immune response (e.g., resulting from administration of a composition comprising a nanoemulsion and an immunogen) provides total or partial immunity to the subject (e.g., from signs, symptoms or conditions of a disease associated with Burkholderia infection (e.g., strep throat, meningitis, bacterial pneumoniae, endocarditis, erysipelas and/or necrotizing fasciitis)). Without being bound to any specific theory, protection and/or immunity from disease (e.g., the ability of a subject's immune system to prevent or attenuate (e.g., suppress) a sign, symptom or condition of disease) after exposure to an immunogenic composition of the present invention is due to adaptive (e.g., acquired) immune responses (e.g., immune responses mediated by B and T cells following exposure to a NE comprising an immunogen of the present invention (e.g., immune responses that exhibit increased specificity and reactivity towards Burkholderia (e.g., Burkholderia cepacia)). Thus, in some embodiments, the compositions and methods of the present invention are used prophylactically or therapeutically to prevent or attenuate a sign, symptom or condition associated with Burkholderia (e.g., Burkholderia cepacia)).
  • In some embodiments, a NE comprising an immunogen (e.g., a Burkholderia (e.g., Burkholderia cepacia) antigen) is administered alone. In some embodiments, a composition comprising a NE and an immunogen (e.g., a Burkholderia (e.g., Burkholderia cepacia) antigen) comprises one or more other agents (e.g., a pharmaceutically acceptable carrier, adjuvant, excipient, and the like). In some embodiments, a composition for stimulating an immune response of the present invention is administered in a manner to induce a humoral immune response. In some embodiments, a composition for stimulating an immune response of the present invention is administered in a manner to induce a cellular (e.g., cytotoxic T lymphocyte) immune response, rather than a humoral response. In some embodiments, a composition comprising a NE and an immunogen of the present invention induces both a cellular and humoral immune response.
  • The present invention is not limited by the isotype, strain or species of Burkholderia (e.g., Burkholderia cepacia) used in a composition comprising a NE and immunogen. Indeed, each Burkholderia (e.g., Burkholderia cepacia) family member alone, or in combination with another family member, may be used to generate a composition comprising a NE and an immunogen (e.g., used to generate an immune response) of the present invention. Exemplary species of Burkholderia are described herein.
  • In some embodiments, the Burkholderia (e.g., Burkholderia cepacia) species utilized is a modified (e.g., genetically modified (e.g., naturally modified via natural selection or modified using recombinant genetic techniques)) species that displays greater pathogenic capacity (e.g., causes more sever Burkholderia—(e.g., Burkholderia cepacia)-induced disease (e.g., comprising enhanced and/or more severe respiratory infection, etc.)). In some embodiments, any one or more members of the genus Burkholderia is utilized in an immunoreactive composition of the invention including but not limited to B. cenocepacia, B. dolosa, B. multivorans, B. ambifaria, B. vietnamiensis, B. ubonensis, B. thailandensis, B. graminis, B. oklahomensis, B. pseudomallei, B. xenovorans, B. phytofirmans, B. phymatum, R. metallidurans, R. eutropha, R. solanacearum.
  • The present invention is not limited by the Burkholderia strain used. Indeed, a variety of Burkholderia strains are contemplated to be useful in the present invention including, but not limited to, classical strains, attenuated strains, non-replicating strains, modified strains (e.g., genetically or mechanically modified strains (e.g., to become more or less virulent)), or other serially diluted strains of Burkholderia. A composition comprising a NE and immunogen may comprise one or more strains of Burkholderia cenocepacia and/or other type of Burkholderia (e.g., Burkholderia multivorans). Additionally, a composition comprising a NE and immunogen may comprise one or more strains of Burkholderia and, in addition, one or more strains of a non-Burkholderia immunogen.
  • In some embodiments, the immunogen may comprise one or more antigens derived from a pathogen (e.g., Burkholderia). For example, in some embodiments, the immunogen is a purified, recombinant, synthetic, or otherwise isolated protein (e.g., added to the NE to generate an immunogenic composition). Similarly, the immunogenic protein may be a derivative, analogue or otherwise modified (e.g., conjugated) form of a protein from a pathogen.
  • The present invention is not limited by the particular formulation of a composition comprising a NE and immunogen of the present invention. Indeed, a composition comprising a NE and immunogen of the present invention may comprise one or more different agents in addition to the NE and immunogen. These agents or cofactors include, but are not limited to, adjuvants, surfactants, additives, buffers, solubilizers, chelators, oils, salts, therapeutic agents, drugs, bioactive agents, antibacterials, and antimicrobial agents (e.g., antibiotics, antivirals, etc.). In some embodiments, a composition comprising a NE and immunogen of the present invention comprises an agent and/or co-factor that enhance the ability of the immunogen to induce an immune response (e.g., an adjuvant). In some preferred embodiments, the presence of one or more co-factors or agents reduces the amount of immunogen required for induction of an immune response (e.g., a protective immune response (e.g., protective immunization)). In some embodiments, the presence of one or more co-factors or agents can be used to skew the immune response towards a cellular (e.g., T cell mediated) or humoral (e.g., antibody mediated) immune response. The present invention is not limited by the type of co-factor or agent used in a therapeutic agent of the present invention.
  • Adjuvants are described in general in Vaccine Design—the Subunit and Adjuvant Approach, edited by Powell and Newman, Plenum Press, New York, 1995. The present invention is not limited by the type of adjuvant utilized (e.g., for use in a composition (e.g., pharmaceutical composition) comprising a NE and immunogen). For example, in some embodiments, suitable adjuvants include an aluminum salt such as aluminum hydroxide gel (alum) or aluminum phosphate. In some embodiments, an adjuvant may be a salt of calcium, iron or zinc, or may be an insoluble suspension of acylated tyrosine, or acylated sugars, cationically or anionically derivatised polysaccharides, or polyphosphazenes.
  • In some embodiments, it is preferred that a composition comprising a NE and immunogen of the present invention comprises one or more adjuvants that induce a Th1-type response. However, in other embodiments, it will be preferred that a composition comprising a NE and immunogen of the present invention comprises one or more adjuvants that induce a Th2-type response.
  • In general, an immune response is generated to an antigen through the interaction of the antigen with the cells of the immune system. Immune responses may be broadly categorized into two categories: humoral and cell mediated immune responses (e.g., traditionally characterized by antibody and cellular effector mechanisms of protection, respectively). These categories of response have been termed Th1-type responses (cell-mediated response), and Th2-type immune responses (humoral response).
  • Stimulation of an immune response can result from a direct or indirect response of a cell or component of the immune system to an intervention (e.g., exposure to an immunogen). Immune responses can be measured in many ways including activation, proliferation or differentiation of cells of the immune system (e.g., B cells, T cells, dendritic cells, APCs, macrophages, NK cells, NKT cells etc.); up-regulated or down-regulated expression of markers and cytokines; stimulation of IgA, IgM, or IgG titer; splenomegaly (including increased spleen cellularity); hyperplasia and mixed cellular infiltrates in various organs. Other responses, cells, and components of the immune system that can be assessed with respect to immune stimulation are known in the art.
  • Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, compositions and methods of the present invention induce expression and secretion of cytokines (e.g., by macrophages, dendritic cells and CD4+ T cells). Modulation of expression of a particular cytokine can occur locally or systemically. It is known that cytokine profiles can determine T cell regulatory and effector functions in immune responses. In some embodiments, Th1-type cytokines can be induced, and thus, the immunostimulatory compositions of the present invention can promote a Th1 type antigen-specific immune response including cytotoxic T-cells (e.g., thereby avoiding unwanted Th2 type immune responses (e.g., generation of Th2 type cytokines (e.g., IL-13) involved in enhancing the severity of disease (e.g., IL-13 induction of mucus formation))).
  • Cytokines play a role in directing the T cell response. Helper (CD4+) T cells orchestrate the immune response of mammals through production of soluble factors that act on other immune system cells, including B and other T cells. Most mature CD4+ T helper cells express one of two cytokine profiles: Th1 or Th2. Th1-type CD4+ T cells secrete IL-2, IL-3, IFN-γ, GM-CSF and high levels of TNF-α. Th2 cells express IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, GM-CSF and low levels of TNF-α. Th1 type cytokines promote both cell-mediated immunity, and humoral immunity that is characterized by immunoglobulin class switching to IgG2a in mice and IgG1 in humans. Th1 responses may also be associated with delayed-type hypersensitivity and autoimmune disease. Th2 type cytokines induce primarily humoral immunity and induce class switching to IgG1 and IgE. The antibody isotypes associated with Th1 responses generally have neutralizing and opsonizing capabilities whereas those associated with Th2 responses are associated more with allergic responses.
  • Several factors have been shown to influence skewing of an immune response towards either a Th1 or Th2 type response. The best characterized regulators are cytokines. IL-12 and IFN-γ are positive Th1 and negative Th2 regulators. IL-12 promotes IFN-γ production, and IFN-γ provides positive feedback for IL-12. IL-4 and IL-10 appear important for the establishment of the Th2 cytokine profile and to down-regulate Th1 cytokine production.
  • Thus, in preferred embodiments, the present invention provides a method of stimulating a Th1-type immune response in a subject comprising administering to a subject a composition comprising a NE and an immunogen. However, in other embodiments, the present invention provides a method of stimulating a Th2-type immune response in a subject (e.g., if balancing of a T cell mediated response is desired) comprising administering to a subject a composition comprising a NE and an immunogen. In further preferred embodiments, adjuvants can be used (e.g., can be co-administered with a composition of the present invention) to skew an immune response toward either a Th1 or Th2 type immune response. For example, adjuvants that induce Th2 or weak Th1 responses include, but are not limited to, alum, saponins, and SB-As4. Adjuvants that induce Th1 responses include but are not limited to MPL, MDP, ISCOMS, IL-12, IFN-γ, and SB-AS2.
  • Several other types of Th1-type immunogens can be used (e.g., as an adjuvant) in compositions and methods of the present invention. These include, but are not limited to, the following. In some embodiments, monophosphoryl lipid A (e.g., in particular 3-de-O-acylated monophosphoryl lipid A (3D-MPL)), is used. 3D-MPL is a well known adjuvant manufactured by Ribi Immunochem, Montana. Chemically it is often supplied as a mixture of 3-de-O-acylated monophosphoryl lipid A with either 4, 5, or 6 acylated chains. In some embodiments, diphosphoryl lipid A, and 3-O-deacylated variants thereof are used. Each of these immunogens can be purified and prepared by methods described in GB 2122204B, hereby incorporated by reference in its entirety. Other purified and synthetic lipopolysaccharides have been described (See, e.g., U.S. Pat. No. 6,005,099 and EP 0 729 473; Hilgers et al., 1986, Int. Arch. Allergy. Immunol., 79(4):392-6; Hilgers et al., 1987, Immunology, 60(1):141-6; and EP 0 549 074, each of which is hereby incorporated by reference in its entirety). In some embodiments, 3D-MPL is used in the form of a particulate formulation (e.g., having a small particle size less than 0.2 μm in diameter, described in EP 0 689 454, hereby incorporated by reference in its entirety).
  • In some embodiments, saponins are used as an immunogen (e.g., Th1-type adjuvant) in a composition of the present invention. Saponins are well known adjuvants (See, e.g., Lacaille-Dubois and Wagner (1996) Phytomedicine vol 2 pp 363-386). Examples of saponins include Quil A (derived from the bark of the South American tree Quillaja Saponaria Molina), and fractions thereof (See, e.g., U.S. Pat. No. 5,057,540; Kensil, Crit. Rev Ther Drug Carrier Syst, 1996, 12 (1-2):1-55; and EP 0 362 279, each of which is hereby incorporated by reference in its entirety). Also contemplated to be useful in the present invention are the haemolytic saponins QS7, QS17, and QS21 (HPLC purified fractions of Quil A; See, e.g., Kensil et al. (1991). J. Immunology 146, 431-437, U.S. Pat. No. 5,057,540; WO 96/33739; WO 96/11711 and EP 0 362 279, each of which is hereby incorporated by reference in its entirety). Also contemplated to be useful are combinations of QS21 and polysorbate or cyclodextrin (See, e.g., WO 99/10008, hereby incorporated by reference in its entirety.
  • In some embodiments, an immunogenic oligonucleotide containing unmethylated CpG dinucleotides (“CpG”) is used as an adjuvant in the present invention. CpG is an abbreviation for cytosine-guanosine dinucleotide motifs present in DNA. CpG is known in the art as being an adjuvant when administered by both systemic and mucosal routes (See, e.g., WO 96/02555, EP 468520, Davis et al., J. Immunol, 1998, 160(2):870-876; McCluskie and Davis, J. Immunol., 1998, 161(9):4463-6; and U.S. Pat. App. No. 20050238660, each of which is hereby incorporated by reference in its entirety). For example, in some embodiments, the immunostimulatory sequence is Purine-Purine-C-G-pyrimidine-pyrimidine; wherein the CG motif is not methylated.
  • Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, the presence of one or more CpG oligonucleotides activate various immune subsets including natural killer cells (which produce IFN-γ) and macrophages. In some embodiments, CpG oligonucleotides are formulated into a composition of the present invention for inducing an immune response. In some embodiments, a free solution of CpG is co-administered together with an antigen (e.g., present within a NE solution (See, e.g., WO 96/02555; hereby incorporated by reference). In some embodiments, a CpG oligonucleotide is covalently conjugated to an antigen (See, e.g., WO 98/16247, hereby incorporated by reference), or formulated with a carrier such as aluminium hydroxide (See, e.g., Brazolot-Millan et al., Proc. Natl. Acad Sci., USA, 1998, 95(26), 15553-8).
  • In some embodiments, adjuvants such as Complete Freunds Adjuvant and Incomplete Freunds Adjuvant, cytokines (e.g., interleukins (e.g., IL-2, IFN-γ, IL-4, etc.), macrophage colony stimulating factor, tumor necrosis factor, etc.), detoxified mutants of a bacterial ADP-ribosylating toxin such as a cholera toxin (CT), a pertussis toxin (PT), or an E. Coli heat-labile toxin (LT), particularly LT-K63 (where lysine is substituted for the wild-type amino acid at position 63) LT-R72 (where arginine is substituted for the wild-type amino acid at position 72), CT-S109 (where serine is substituted for the wild-type amino acid at position 109), and PT-K9/G129 (where lysine is substituted for the wild-type amino acid at position 9 and glycine substituted at position 129) (See, e.g., WO93/13202 and WO92/19265, each of which is hereby incorporated by reference), and other immunogenic substances (e.g., that enhance the effectiveness of a composition of the present invention) are used with a composition comprising a NE and immunogen of the present invention.
  • Additional examples of adjuvants that find use in the present invention include poly(di(carboxylatophenoxy)phosphazene (PCPP polymer; Virus Research Institute, USA); derivatives of lipopolysaccharides such as monophosphoryl lipid A (MPL; Ribi ImmunoChem Research, Inc., Hamilton, Mont.), muramyl dipeptide (MDP; Ribi) and threonyl-muramyl dipeptide (t-MDP; Ribi); OM-174 (a glucosamine disaccharide related to lipid A; OM Pharma SA, Meyrin, Switzerland); and Leishmania elongation factor (a purified Leishmania protein; Corixa Corporation, Seattle, Wash.).
  • Adjuvants may be added to a composition comprising a NE and an immunogen, or, the adjuvant may be formulated with carriers, for example liposomes, or metallic salts (e.g., aluminium salts (e.g., aluminium hydroxide)) prior to combining with or co-administration with a composition comprising a NE and an immunogen.
  • In some embodiments, a composition comprising a NE and an immunogen comprises a single adjuvant. In other embodiments, a composition comprising a NE and an immunogen comprises two or more adjuvants (See, e.g., WO 94/00153; WO 95/17210; WO 96/33739; WO 98/56414; WO 99/12565; WO 99/11241; and WO 94/00153, each of which is hereby incorporated by reference in its entirety).
  • In some embodiments, a composition comprising a NE and an immunogen of the present invention comprises one or more mucoadhesives (See, e.g., U.S. Pat. App. No. 20050281843, hereby incorporated by reference in its entirety). The present invention is not limited by the type of mucoadhesive utilized. Indeed, a variety of mucoadhesives are contemplated to be useful in the present invention including, but not limited to, cross-linked derivatives of poly(acrylic acid) (e.g., carbopol and polycarbophil), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides (e.g., alginate and chitosan), hydroxypropyl methylcellulose, lectins, fimbrial proteins, and carboxymethylcellulose. Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, use of a mucoadhesive (e.g., in a composition comprising a NE and immunogen) enhances induction of an immune response in a subject (e.g., administered a composition of the present invention) due to an increase in duration and/or amount of exposure to an immunogen that a subject experiences when a mucoadhesive is used compared to the duration and/or amount of exposure to an immunogen in the absence of using the mucoadhesive.
  • In some embodiments, a composition of the present invention may comprise sterile aqueous preparations. Acceptable vehicles and solvents include, but are not limited to, water, Ringer's solution, phosphate buffered saline and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed mineral or non-mineral oil may be employed including synthetic mono-ordi-glycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Carrier formulations suitable for mucosal, subcutaneous, intramuscular, intraperitoneal, intravenous, or administration via other routes may be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • A composition comprising a NE and an immunogen of the present invention can be used therapeutically (e.g., to enhance an immune response) or as a prophylactic (e.g., for immunization (e.g., to prevent signs or symptoms of disease)). A composition comprising a NE and an immunogen of the present invention can be administered to a subject via a number of different delivery routes and methods.
  • For example, an immunogenic compositions of the invention can be administered to a subject (e.g., mucosally (e.g., nasal mucosa, vaginal mucosa, etc.)) by multiple methods, including, but not limited to: being suspended in a solution and applied to a surface; being suspended in a solution and sprayed onto a surface using a spray applicator; being mixed with a mucoadhesive and applied (e.g., sprayed or wiped) onto a surface (e.g., mucosal surface); being placed on or impregnated onto a nasal and/or vaginal applicator and applied; being applied by a controlled-release mechanism; being applied as a liposome; or being applied on a polymer.
  • In some preferred embodiments, immunogenic compositions described herein are administered mucosally (e.g., using standard techniques; See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995 (e.g., for mucosal delivery techniques, including intranasal, pulmonary, vaginal and rectal techniques), as well as European Publication No. 517,565 and Illum et al., J. Controlled Rel., 1994, 29:133-141 (e.g., for techniques of intranasal administration), each of which is hereby incorporated by reference in its entirety). Alternatively, immunogenic compositions described herein are administered dermally or transdermally, using standard techniques (See, e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th edition, 1995). The present invention is not limited by the route of administration.
  • Mucosal vaccination, such as intranasal vaccination, may induce mucosal immunity not only in the nasal mucosa, but also in distant mucosal sites such as the genital mucosa (See, e.g., Mestecky, Journal of Clinical Immunology, 7:265-276, 1987). In addition to inducing mucosal immune responses, mucosal vaccination also induces systemic immunity (See, e.g., Example 10). In some embodiments, non-parenteral administration (e.g., mucosal administration of vaccines) provides an efficient and convenient way to boost systemic immunity (e.g., induced by parenteral or mucosal vaccination (e.g., in cases where multiple boosts are used to sustain a vigorous systemic immunity)).
  • In some embodiments, a composition comprising a NE and an immunogen of the present invention may be used to protect or treat a subject susceptible to, or suffering from, disease by means of administering a composition of the present invention via a mucosal route (e.g., an oral/alimentary or nasal route). Alternative mucosal routes include intravaginal and intra-rectal routes. In preferred embodiments of the present invention, a nasal route of administration is used, termed “intranasal administration” or “intranasal vaccination” herein. Methods of intranasal vaccination are well known in the art, including the administration of a droplet or spray form of the vaccine into the nasopharynx of a subject to be immunized. In some embodiments, a nebulized or aerosolized composition comprising a NE and immunogen is provided. Enteric formulations such as gastro resistant capsules for oral administration, suppositories for rectal or vaginal administration also form part of this invention. Immunogenic compositions described herein may also be administered via the oral route. Under these circumstances, a composition comprising a NE and an immunogen may comprise a pharmaceutically acceptable excipient and/or include alkaline buffers, or enteric capsules. Formulations for nasal delivery may include those with dextran or cyclodextran and saponin as an adjuvant.
  • Immunogenic compositions described herein may also be administered via a vaginal route. In such cases, a composition comprising a NE and an immunogen may comprise pharmaceutically acceptable excipients and/or emulsifiers, polymers (e.g., CARBOPOL), and other known stabilizers of vaginal creams and suppositories. In some embodiments, compositions of the present invention are administered via a rectal route. In such cases, a composition comprising a NE and an immunogen may comprise excipients and/or waxes and polymers known in the art for forming rectal suppositories.
  • In some embodiments, the same route of administration (e.g., mucosal administration) is chosen for both a priming and boosting vaccination. In some embodiments, multiple routes of administration are utilized (e.g., at the same time, or, alternatively, sequentially) in order to stimulate an immune response (e.g., using an immunogenic compositions described herein).
  • For example, in some embodiments, a composition comprising a NE and an immunogen is administered to a mucosal surface of a subject in either a priming or boosting vaccination regime. Alternatively, in some embodiments, a composition comprising a NE and an immunogen is administered systemically in either a priming or boosting vaccination regime. In some embodiments, a composition comprising a NE and an immunogen is administered to a subject in a priming vaccination regimen via mucosal administration and a boosting regimen via systemic administration. In some embodiments, a composition comprising a NE and an immunogen is administered to a subject in a priming vaccination regimen via systemic administration and a boosting regimen via mucosal administration. Examples of systemic routes of administration include, but are not limited to, a parenteral, intramuscular, intradermal, transdermal, subcutaneous, intraperitoneal or intravenous administration. A composition comprising a NE and an immunogen may be used for both prophylactic and therapeutic purposes.
  • In some embodiments, immunogenic compositions described herein are administered by pulmonary delivery. For example, immunogenic compositions described herein can be delivered to the lungs of a subject (e.g., a human) via inhalation (e.g., thereby traversing across the lung epithelial lining to the blood stream (See, e.g., Adjei, et al. Pharmaceutical Research 1990; 7:565-569; Adjei, et al. Int. J. Pharmaceutics 1990; 63:135-144; Braquet, et al. J. Cardiovascular Pharmacology 1989 143-146; Hubbard, et al. (1989) Annals of Internal Medicine, Vol. III, pp. 206-212; Smith, et al. J. Clin. Invest. 1989; 84:1145-1146; Oswein, et al. “Aerosolization of Proteins”, 1990; Proceedings of Symposium on Respiratory Drug Delivery II Keystone, Colo.; Debs, et al. J. Immunol. 1988; 140:3482-3488; and U.S. Pat. No. 5,284,656 to Platz, et al, each of which are hereby incorporated by reference in its entirety). A method and composition for pulmonary delivery of drugs for systemic effect is described in U.S. Pat. No. 5,451,569 to Wong, et al., hereby incorporated by reference; See also U.S. Pat. No. 6,651,655 to Licalsi et al., hereby incorporated by reference in its entirety)). Further contemplated for use in the practice of delivery of immunogenic compositions described herein are the wide range of mechanical devices designed for pulmonary and/or nasal mucosal delivery of pharmaceutical agents described herein
  • Thus, in some embodiments, an immunogenic composition described herein may be used to protect and/or treat a subject susceptible to, or suffering from, a disease by means of administering a compositions comprising a NE and an immunogen by mucosal, intramuscular, intraperitoneal, intradermal, transdermal, pulmonary, intravenous, subcutaneous or other route of administration described herein under conditions that induce an immunogen-specific immune response in the subject. Methods of systemic administration of the vaccine preparations may include conventional syringes and needles, or devices designed for ballistic delivery of solid vaccines (See, e.g., WO 99/27961, hereby incorporated by reference), or needleless pressure liquid jet device (See, e.g., U.S. Pat. No. 4,596,556; U.S. Pat. No. 5,993,412, each of which are hereby incorporated by reference), or transdermal patches (See, e.g., WO 97/48440; WO 98/28037, each of which are hereby incorporated by reference). The present invention may also be used to enhance the immunogenicity of antigens applied to the skin (transdermal or transcutaneous delivery, See, e.g., WO 98/20734; WO 98/28037, each of which are hereby incorporated by reference). Thus, in some embodiments, the present invention provides a delivery device for systemic administration, pre-filled with the vaccine composition of the present invention.
  • The present invention is not limited by the type of subject administered (e.g., in order to stimulate an immune response (e.g., in order to generate protective immunity (e.g., mucosal and/or systemic immunity))) a composition of the present invention. Indeed, a wide variety of subjects are contemplated to be benefited from administration of a composition of the present invention. In preferred embodiments, the subject is a human. In some embodiments, human subjects are of any age (e.g., adults, children, infants, etc.) that have been or are likely to become exposed to a microorganism (e.g., bacteria of the genus Burkholderia). In some embodiments, the human subjects are subjects that are more likely to suffer from opportunistic infection (e.g., a subject with CF or an immune suppressed subject (e.g., a subject with human immunodeficiency virus)). In some embodiments, the subjects are non-human mammals (e.g., pigs, cattle, goats, horses, sheep, or other livestock; or mice, rats, rabbits or other animal). In some embodiments, compositions and methods of the present invention are utilized in research settings (e.g., with research animals). In some embodiments, the present invention provides a method to elicit an immune response (e.g., protective immune response) in infants (e.g., from about 0-2 years old) by administering to the infant a safe and effective amount of an immunogenic composition of the invention (e.g., a pediatric vaccine). Further embodiments of the invention include the provision of the immunogenic Burkholderia nanoemulsion compositions of the invention for use in medicine and the use of the Burkholderia nanoemulsion compositions of the invention in the manufacture of a medicament for the prevention (or treatment) of disease caused by Burkholderia. In yet another embodiment, the present invention is provides a method to elicit an immune response (e.g., a protective immune response) in the elderly population (e.g., in a subject 50 years or over in age, typically over 55 years and more generally over 60 years) by administering a safe and effective amount of an immunogenic composition of the invention.
  • Immunogenic compositions described herein may be formulated for administration by any route, such as mucosal, oral, topical, parenteral or other route described herein. The compositions may be in any one or more different forms including, but not limited to, tablets, capsules, powders, granules, lozenges, foams, creams or liquid preparations.
  • Topical formulations may be presented as, for instance, ointments, creams or lotions, foams, and aerosols, and may contain appropriate conventional additives such as preservatives, solvents (e.g., to assist penetration), and emollients in ointments and creams.
  • Topical formulations may also include agents that enhance penetration of the active ingredients through the skin. Exemplary agents include a binary combination of N-(hydroxyethyl)pyrrolidone and a cell-envelope disordering compound, a sugar ester in combination with a sulfoxide or phosphine oxide, and sucrose monooleate, decyl methyl sulfoxide, and alcohol.
  • Other exemplary materials that increase skin penetration include surfactants or wetting agents including, but not limited to, polyoxyethylene sorbitan mono-oleoate (Polysorbate 80); sorbitan mono-oleate (Span 80); p-isooctyl polyoxyethylene-phenol polymer (Triton WR-1330); polyoxyethylene sorbitan tri-oleate (Tween 85); dioctyl sodium sulfosuccinate; and sodium sarcosinate (Sarcosyl NL-97); and other pharmaceutically acceptable surfactants.
  • In certain embodiments of the invention, compositions may further comprise one or more alcohols, zinc-containing compounds, emollients, humectants, thickening and/or gelling agents, neutralizing agents, and surfactants. Water used in the formulations is preferably deionized water having a neutral pH. Additional additives in the topical formulations include, but are not limited to, silicone fluids, dyes, fragrances, pH adjusters, and vitamins.
  • Topical formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation. The ointment base can comprise one or more of petrolatum, mineral oil, ceresin, lanolin alcohol, panthenol, glycerin, bisabolol, cocoa butter and the like.
  • Immunogenic compositions described herein may additionally contain other adjunct components conventionally found in pharmaceutical compositions. Thus, for example, the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipuritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, preferably do not unduly interfere with the biological activities of the immunogenic compositions described herein. The formulations can be sterilized and, if desired, mixed with auxiliary agents (e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like) that do not deleteriously interact with the NE and immunogen of the formulation. In some embodiments, immunogenic compositions described herein are administered in the form of a pharmaceutically acceptable salt. When used the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include, but are not limited to, acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v). Suitable preservatives may include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • In some embodiments, a composition comprising a NE and an immunogen is co-administered with one or more antibiotics. For example, one or more antibiotics may be administered with, before and/or after administration of a composition comprising a NE and an immunogen. The present invention is not limited by the type of antibiotic co-administered. Indeed, a variety of antibiotics may be co-administered including, but not limited to, □-lactam antibiotics, penicillins (such as natural penicillins, aminopenicillins, penicillinase-resistant penicillins, carboxy penicillins, ureido penicillins), cephalosporins (first generation, second generation, and third generation cephalosporins), and other β-lactams (such as imipenem, monobactams), β-lactamase inhibitors, vancomycin, aminoglycosides and spectinomycin, tetracyclines, chloramphenicol, erythromycin, lincomycin, clindamycin, rifampin, metronidazole, polymyxins, doxycycline, quinolones (e.g., ciprofloxacin), sulfonamides, trimethoprim, and quinolines.
  • There are an enormous amount of antimicrobial agents currently available for use in treating bacterial, fungal and viral infections. For a comprehensive treatise on the general classes of such drugs and their mechanisms of action, the skilled artisan is referred to Goodman & Gilman's “The Pharmacological Basis of Therapeutics” Eds. Hardman et al., 9th Edition, Pub. McGraw Hill, chapters 43 through 50, 1996, (herein incorporated by reference in its entirety). Generally, these agents include agents that inhibit cell wall synthesis (e.g., penicillins, cephalosporins, cycloserine, vancomycin, bacitracin); and the imidazole antifungal agents (e.g., miconazole, ketoconazole and clotrimazole); agents that act directly to disrupt the cell membrane of the microorganism (e.g., detergents such as polmyxin and colistimethate and the antifungals nystatin and amphotericin B); agents that affect the ribosomal subunits to inhibit protein synthesis (e.g., chloramphenicol, the tetracyclines, erythromycin and clindamycin); agents that alter protein synthesis and lead to cell death (e.g., aminoglycosides); agents that affect nucleic acid metabolism (e.g., the rifamycins and the quinolones); the antimetabolites (e.g., trimethoprim and sulfonamides); and the nucleic acid analogues such as zidovudine, gangcyclovir, vidarabine, and acyclovir which act to inhibit viral enzymes essential for DNA synthesis. Various combinations of antimicrobials may be employed.
  • The present invention also includes methods involving co-administration of a composition comprising a NE and an immunogen with one or more additional active and/or immunostimulatory agents (e.g., a composition comprising a NE and a different immunogen, an antibiotic, anti-oxidant, etc.). Indeed, it is a further aspect of this invention to provide methods for enhancing prior art immunostimulatory methods (e.g., immunization methods) and/or pharmaceutical compositions by co-administering a composition of the present invention. In co-administration procedures, the agents may be administered concurrently or sequentially. In one embodiment, the compositions described herein are administered prior to the other active agent(s). The pharmaceutical formulations and modes of administration may be any of those described herein. In addition, the two or more co-administered agents may each be administered using different modes (e.g., routes) or different formulations. The additional agents to be co-administered (e.g., antibiotics, adjuvants, etc.) can be any of the well-known agents in the art, including, but not limited to, those that are currently in clinical use.
  • In some embodiments, a composition comprising a NE and immunogen is administered to a subject via more than one route. For example, a subject that would benefit from having a protective immune response (e.g., immunity) towards a pathogenic microorganism may benefit from receiving mucosal administration (e.g., nasal administration or other mucosal routes described herein) and, additionally, receiving one or more other routes of administration (e.g., parenteral or pulmonary administration (e.g., via a nebulizer, inhaler, or other methods described herein). In some preferred embodiments, administration via mucosal route is sufficient to induce both mucosal as well as systemic immunity towards an immunogen or organism from which the immunogen is derived. In other embodiments, administration via multiple routes serves to provide both mucosal and systemic immunity. Thus, although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, it is contemplated that a subject administered a composition of the present invention via multiple routes of administration (e.g., immunization (e.g., mucosal as well as airway or parenteral administration of a composition comprising a NE and immunogen of the present invention) may have a stronger immune response to an immunogen than a subject administered a composition via just one route.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the compositions, increasing convenience to the subject and a physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer based systems such as poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109, hereby incorporated by reference. Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like. Specific examples include, but are not limited to: (a) erosional systems in which an agent of the invention is contained in a form within a matrix such as those described in U.S. Pat. Nos. 4,452,775, 4,675,189, and 5,736,152, each of which is hereby incorporated by reference and (b) diffusional systems in which an active component permeates at a controlled rate from a polymer such as described in U.S. Pat. Nos. 3,854,480, 5,133,974 and 5,407,686, each of which is hereby incorporated by reference. In addition, pump-based hardware delivery systems can be used, some of which are adapted for implantation.
  • In preferred embodiments, a composition comprising a NE and an immunogen of the present invention comprises a suitable amount of the immunogen to induce an immune response in a subject when administered to the subject. In preferred embodiments, the immune response is sufficient to provide the subject protection (e.g., immune protection) against a subsequent exposure to the immunogen or the microorganism (e.g., bacteria of the genus Burkholderia) from which the immunogen was derived. The present invention is not limited by the amount of immunogen used. In some preferred embodiments, the amount of immunogen (e.g., Burkholderia bacteria (e.g., Burkholderia cepacia) or one or more component parts thereof) in a composition comprising a NE and immunogen (e.g., for use as an immunization dose) is selected as that amount which induces an immunoprotective response without significant, adverse side effects. The amount will vary depending upon which specific immunogen or combination thereof is/are employed, and can vary from subject to subject, depending on a number of factors including, but not limited to, the species, age and general condition (e.g., health) of the subject, and the mode of administration.
  • In some embodiments, each dose (e.g., of a composition comprising a NE and an immunogen (e.g., administered to a subject to induce an immune response (e.g., a protective immune response (e.g., protective immunity))) comprises about 0.05-5000 μg of immunogen (e.g., recombinant and/or purified Burkholderia OMP protein). In some embodiments, each dose (e.g., of a composition comprising a NE and an immunogen (e.g., administered to a subject to induce an immune response (e.g., a protective immune response (e.g., protective immunity))) comprises about 0.05-5000 μg of the immunogen (e.g., recombinant and/or purified Burkholderia OMP protein) and comprises about 0.05-5000 μg of another immunogen (e.g., recombinant and/or purified protein, adjuvant (e.g., cholera toxin), etc.). In some embodiments, each dose will comprise 1-500 μg, in some embodiments, each dose will comprise 350-750 μg, in some embodiments, each dose will comprise 50-200 μg, in some embodiments, each dose will comprise 25-75 μg of immunogen (e.g., recombinant and/or purified protein (e.g., Burkholderia antigen). In some embodiments, each dose comprises an amount of the immunogen sufficient to generate an immune response. An effective amount of the immunogen in a dose need not be quantified, as long as the amount of immunogen generates an immune response in a subject when administered to the subject.
  • In some embodiments, it is expected that each dose (e.g., of a composition comprising a NE and an immunogen (e.g., administered to a subject to induce and immune response)) is from 0.001 to 15% or more (e.g., 0.001-10%, 0.5-5%, 1-3%, 2%, 6%, 10%, 15% or more) by weight immunogen (e.g., neutralized bacteria, or recombinant and/or purified protein). In some embodiments, an initial or prime administration dose contains more immunogen than a subsequent boost dose
  • In some embodiments, a composition comprising a NE and an immunogen of the present invention is formulated in a concentrated dose that can be diluted prior to administration to a subject. For example, dilutions of a concentrated composition may be administered to a subject such that the subject receives any one or more of the specific dosages provided herein. In some embodiments, dilution of a concentrated composition may be made such that a subject is administered (e.g., in a single dose) a composition comprising 0.5-50% of the NE and immunogen present in the concentrated composition. In some preferred embodiments, a subject is administered in a single dose a composition comprising 1% of the NE and immunogen present in the concentrated composition. Concentrated compositions are contemplated to be useful in a setting in which large numbers of subjects may be administered a composition of the present invention (e.g., an immunization clinic, hospital, school, etc.). In some embodiments, a composition comprising a NE and an immunogen of the present invention (e.g., a concentrated composition) is stable at room temperature for more than 1 week, in some embodiments for more than 2 weeks, in some embodiments for more than 3 weeks, in some embodiments for more than 4 weeks, in some embodiments for more than 5 weeks, and in some embodiments for more than 6 weeks.
  • Generally, the emulsion compositions of the invention will comprise at least 0.001% to 100%, preferably 0.01 to 90%, of emulsion per ml of liquid composition. It is envisioned that the formulations may comprise about 0.001%, about 0.0025%, about 0.005%, about 0.0075%, about 0.01%, about 0.025%, about 0.05%, about 0.075%, about 0.1%, about 0.25%, about 0.5%, about 1.0%, about 2.5%, about 5%, about 7.5%, about 10%, about 12.5%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or about 98% of emulsion per ml of liquid composition. It should be understood that a range between any two figures listed above is specifically contemplated to be encompassed within the metes and bounds of the present invention. Some variation in dosage will necessarily occur depending on the condition of the specific pathogen and the subject being immunized.
  • In some embodiments, following an initial administration of a composition of the present invention (e.g., an initial vaccination), a subject may receive one or more boost administrations (e.g., around 2 weeks, around 3 weeks, around 4 weeks, around 5 weeks, around 6 weeks, around 7 weeks, around 8 weeks, around 10 weeks, around 3 months, around 4 months, around 6 months, around 9 months, around 1 year, around 2 years, around 3 years, around 5 years, around 10 years) subsequent to a first, second, third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, and/or more than tenth administration. Although an understanding of the mechanism is not necessary to practice the present invention and the present invention is not limited to any particular mechanism of action, in some embodiments, reintroduction of an immunogen in a boost dose enables vigorous systemic immunity in a subject. The boost can be with the same formulation given for the primary immune response, or can be with a different formulation that contains the immunogen. The dosage regimen will also, at least in part, be determined by the need of the subject and be dependent on the judgment of a practitioner.
  • Dosage units may be proportionately increased or decreased based on several factors including, but not limited to, the weight, age, and health status of the subject. In addition, dosage units may be increased or decreased for subsequent administrations (e.g., boost administrations).
  • It is contemplated that the compositions and methods of the present invention will find use in various settings, including research settings. For example, compositions and methods of the present invention also find use in studies of the immune system (e.g., characterization of adaptive immune responses (e.g., protective immune responses (e.g., mucosal or systemic immunity))). Uses of the compositions and methods provided by the present invention encompass human and non-human subjects and samples from those subjects, and also encompass research applications using these subjects. Compositions and methods of the present invention are also useful in studying and optimizing nanoemulsions, immunogens, and other components and for screening for new components. Thus, it is not intended that the present invention be limited to any particular subject and/or application setting.
  • The formulations can be tested in vivo in a number of animal models developed for the study of mucosal and other routes of delivery. As is readily apparent, the compositions of the present invention are useful for preventing and/or treating a wide variety of diseases and infections caused by viruses, bacteria, parasites, and fungi, as well as for eliciting an immune response against a variety of antigens. Not only can the compositions be used prophylactically or therapeutically, as described above, the compositions can also be used in order to prepare antibodies, both polyclonal and monoclonal (e.g., for diagnostic purposes), as well as for immunopurification of an antigen of interest. If polyclonal antibodies are desired, a selected mammal, (e.g., mouse, rabbit, goat, horse, etc.) can be immunized with the compositions of the present invention. The animal is usually boosted 2-6 weeks later with one or more—administrations of the antigen. Polyclonal antisera can then be obtained from the immunized animal and used according to known procedures (See, e.g., Jurgens et al., J. Chrom. 1985, 348:363-370).
  • In some embodiments, the present invention provides a kit comprising a composition comprising a NE and an immunogen. In some embodiments, the kit further provides a device for administering the composition. The present invention is not limited by the type of device included in the kit. In some embodiments, the device is configured for nasal application of the composition of the present invention (e.g., a nasal applicator (e.g., a syringe) or nasal inhaler or nasal mister). In some embodiments, a kit comprises a composition comprising a NE and an immunogen in a concentrated form (e.g., that can be diluted prior to administration to a subject).
  • In some embodiments, all kit components are present within a single container (e.g., vial or tube). In some embodiments, each kit component is located in a single container (e.g., vial or tube). In some embodiments, one or more kit component are located in a single container (e.g., vial or tube) with other components of the same kit being located in a separate container (e.g., vial or tube). In some embodiments, a kit comprises a buffer. In some embodiments, the kit further comprises instructions for use.
  • EXPERIMENTAL
  • The following examples are provided in order to demonstrate and further illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
  • Example 1 In Vivo Toxicity Studies
  • A nanoemulsion composition of the present invention was tested to determine if pulmonary administration of the composition to a subject would elicit any histological changes and/or pathology.
  • A solution comprising 20% nanoemulsion (W805EC) and 0.1% EDTA was administered via a nebulizer. A custom murine nose only nebulization chamber with PARI LC nebulizer (Midlothian, Va.) and compressor was used. A 0.9% NaCl solution was used as a control. Using the nebulizer, mice were administered nebulized nanoemulsion+EDTA or NaCl solution for 10 minutes. Twenty-four hours after administration, mice were sacrificed and physical properties and histology assessed.
  • Upon examination, there was an absence of histological changes indicating the absence of toxicity upon administration of nebulized nanoemulsion. Physical findings were normal.
  • Example 2 Killing Assays Utilizing Nanoemulsion Compositions and Bacteria Found in the Respiratory Tract
  • It was determined whether a composition comprising nanoemulsion (W805EC) alone or in combination with EDTA and/or the presence of a hypertonic salt solution would be able to attenuate growth and/or kill bacteria found in the respiratory tract.
  • An overnight culture of Burkholderia cepacia or Pseudomonas aeruginosa was started in 6 ml of cation adjusted Mueller Hinton Broth (MHB) from a frozen bacterial stock at −80° C. The culture was incubated in a shaking incubator at 37° C. The following day, bacteria were brought to logarithmic growth phase by back diluting the overnight culture 1:4 with fresh MHB. Back diluted culture was incubated at 37° C. in the shaking incubator until the OD600 reached between 0.40 to 0.45. One ml of the culture was spun down at 3500 to 4000 rpm for 15 minutes. The bacterial pellet was resuspended in 1 ml of sterile 2×PBS, 12% or 14% saline. Appropriate dilutions were done with the same solutions to get desired numbers of bacteria in 50 μl (OD600 of 0.5 approximately =109 bacteria). Dilutions of the starting bacterial suspensions were plated onto Luria Bertani (LB) agar plates to estimate the starting bacterial counts. Double the concentrations of the desired final nanoemulsion and nanoemulsion with EDTA concentrations were made with sterile milliQ water. Fifty micro liters of the nanoemulsion was mixed with 50 μl of the bacteria and vortexed to mix the reaction. The mixture was incubated at 37° C. for desired time intervals. Following incubation, the reaction was diluted with 500 μl of 1×PBS and vortexed to mix the contents. The contents were centrifuged at 4000 rpm for 15 minutes to pellet the bacteria and separate the nanoemulsion. Supernatant containing the nanoemulsion was removed and the bacterial pellet resuspended in 1×PBS. Undiluted or dilutions of the resuspended bacterial pellet was plated onto LB agar plates for colony count. Log killing of the bacteria were calculated from ratio with the starting numbers.
  • The following killing assays were performed:
  • Killing of Burkholderia cepacia in PBS by 20% Nanemulsion (NE) alone or with 20 mM EDTA in 10 minutes (See FIG. 1);
  • Killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA within 20 minutes (See FIG. 2);
  • Killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA within 40 minutes (See FIG. 3);
  • Killing of B. cepacia in PBS by 20% NE alone or with 20 mM EDTA within 40 or 60 minutes (See FIG. 4);
  • Killing of B. cepacia in hypertonic saline (6% NaCl) at 15 and 30 minutes by NE alone and NE with EDTA (See FIG. 5);
  • Killing of B. cepacia and P. aeruginosa in 7% NaCl within 15 min (See FIG. 6); and
  • Killing of B. cepacia and P. aeruginosa (Mixed-culture) in 7% NaCl within 15 minutes (See FIG. 7).
  • Nanoemulsion alone or in combination with EDTA (e.g., 10-20 mM EDTA) was able to achieve complete killing of 106 bacteria in PBS in 60 minutes. In the presence of hypertonic saline (e.g., 6-7% NaCl), the killing ability of the nanoemulsion was strikingly enhanced, achieving complete killing within 15 minutes while in the presence of 20 mM EDTA. Also, nanoemulsions comprising a lower concentration of EDTA were able to achieve complete killing of bacteria in 30 minutes in the presence of hypertonic saline. Thus, the present invention provides that a nanoemulsion composition comprising EDTA can be used to kill (e.g., completely) bacteria over a short time period (e.g., <60 minutes). Moreover, the present invention provides that nanoemulsion with hypertonic saline and EDTA can be used to kill bacteria over even shorter time periods (e.g., <30 minutes, <15 minutes) and that the concentration of EDTA and hypertonic saline solution alter the pace at which the nanoemulsion is able to eradicate the bacteria. The present invention also demonstrates that compositions of the present invention are able to eradicate a mixed population of bacteria.
  • Example 3 Nanoemulsion Killing of Cystic Fibrosis (CF) Related Bacteria Including Multi-Drug Resistant Strains
  • Materials and Methods.
  • Bacterial strains and culture conditions. One hundred fifty isolates were analyzed including 75 Burkholderia isolates and 75 isolates belonging to other CF-relevant species, including Pseudomonas aeruginosa, Achromobacter xylosoxidans, Stenotrophomonas maltophilia, Acinetobacter species, Pandoraea species (P. apista, P. pnomenusa, P. pulmonicola, P. norimburgensis, and P. sputorum), and Ralstonia species (R. mannitolilytica and R. pickettii). One hundred forty-five clinical isolates were obtained from the Burkholderia cepacia Research Laboratory and Repository (BcRLR, University of Michigan, Ann Arbor, Mich.). These were recovered from 142 individuals between September 1997 and October 2007 and were referred to the BcRLR from 62 CF treatment centers in the U.S. for analysis. The remaining five strains included environmental isolates B. multivorans ATCC 17616 (American Type Culture Collection, Manassas, Va.), P. norimburgensis LMG 18379T (BCCM/LMG Bacteria Collection, Laboratorium voor Micrbiologie Gent, Universiteit Gent, Ghent, Belgium) and B. pyrrocinia HI3642 (BcRLR collection), and the clinical type strains B. cenocepacia LMG 16656T (aka J2315) and P. pulmonicola LMG 18106T. One hundred thirty four (91%) of the 147 clinical isolates were recovered from persons with CF; 114 (78%) of these were from sputum culture, with the remainder from throat swab (n=17), endotracheal suction/tracheal aspiration (n=5), blood (n=5), bronchial lavage (n=3), and one each from maxillary sinus, peritoneal cavity, and epiglottis. Forty nine (33%) of the 150 isolates were defined as multi-drug resistant (resistant to all drugs tested in two of three antibiotic classes: lactams including carbapenems, aminoglycosides, and quinolones; See, e.g., Taccetti et al., Eur J Epidemiol. 1999 January; 15(1):85-8) based on susceptibility testing performed at the referring microbiology laboratory; 20 (41%) of these were panresistant. Seventy two (48%) of the remaining isolates were susceptible to at least one antibiotic, and susceptibility testing results were unavailable for 29 (19%) isolates. All isolates were identified to the species level at the BcRLR by polyphasic analyses using phenotypic and genotypic assays as described (See, e.g., Reik et al., J Clin Microbiol. 2005 June; 43(6):2926-8). The exception was Acinetobacter, which was identified only to the genus level due to lack of definitive species-specific assays. All isolates were also subjected to repetitive extragenic element-PCR (rep-PCR) typing using the BOX A1R primer as previously described (See, e.g., Coenye et al., J Clin Microbiol. 2002 September; 40(9):3300-7) to ensure that all 150 isolates included in the test panel were genotypically distinct. Bacteria were stored at −80° C. in skim milk or Lauria-Bertani (LB) broth with 15% glycerol and recovered from frozen stock overnight at 37° C. on Mueller-Hinton (MH) agar.
  • Nanoemulsion. Nanoemulsion P4075EC, described herein, was manufactured by NANOBIO Corp. (Ann Arbor, Mich.). P4075EC droplets had a mean particle diameter of 400 nm. Surfactants and food substances utilized in the manufacture of P4075EC were ‘Generally Recognized as Safe’ (GRAS) by the FDA, and manufactured in accordance with Good Manufacturing Practices (GMP). The concentration of CPC was used as a surrogate for the amount of P4075EC used experimentally. P4075EC was stable for no less than 12 months at 40° C.
  • Susceptibility testing. Because P4075EC is opaque, the MICs of this compound for test bacteria were determined by using a modification of the Clinical and Laboratory Standards Institute (CLSI)-approved microtiter serial dilution method (See, e.g., Clinical and Laboratory Standards Institute. 2006. Approved standard M7-A7, seventh edition. Clinical and Laboratory Standards Institute, Wayne, Pa.). P4075EC was diluted to a concentration of 2 mg/ml (of CPC) in MH broth supplemented with 7% NaCl and 20 mM EDTA. Serial two-fold dilutions of this preparation were made in unsupplemented MH broth and aliquoted into 96-well flat bottom microtiter plates (100 μl/well). Bacteria from overnight growth on MH agar were suspended in MH broth to a 0.5 McFarland turbidity standard (absorbance of 0.08-0.13 at 625 nm), further diluted 1:100 in MH broth, and added (5 μl/well) to the P4075EC serial dilution wells. Appropriate controls, including wells with bacteria but no P4075EC and wells with P4075EC dilutions but no bacteria, were included on each plate. Microtiter plates were shaken briefly, and 1 μl was removed from wells containing bacteria but no P4075EC, diluted in 1 ml of MH broth, plated on MH agar (100 μl), and incubated for 24-48 h at 37° C. to determine bacterial concentrations of initial inoculums. Microtiter plates were then incubated at 37° C. without shaking. To determine MBCs, 10 μl were removed from each well after overnight growth, spotted on MH agar, and incubated at 37° C. Colonies were enumerated 24 h later, and MBCs were recorded as the P4075EC concentration with a 3 log decrease in CFU/ml compared to the initial inoculum. To determine MICs, 10 μl of resazurin (R&D SYSTEMS, Minneapolis, Minn.) were added to each well and microtiter plates were shaken briefly, covered with foil, and incubated at 37° C. without shaking. Wells were visually inspected the next day, and the MIC was recorded as the lowest concentration of P4075EC remaining a blue color. MIC results were further quantified by recording fluorescence on a spectrofluorometer at 560 nm excitation/590 nm emission.
  • Biofilm growth and susceptibility testing. To identify biofilm-forming isolates, bacteria were grown overnight in tryptic soy broth, adjusted to a 0.5 McFarland turbidity standard, and further diluted 1:10 in MH broth, and 100 μl were seeded in triplicate in internal wells of 96-well flat bottom microtiter plates. Negative control wells without bacteria were included. To minimize evaporation, the remaining wells were filled with MH broth, and the plate was wrapped with plastic wrap before incubating for 48 h at 37° C. without shaking. Wells were gently washed twice with phosphate buffered saline (PBS; pH 7.4), dried for 2 h at 37° C., and then stained with 1% crystal violet in water for 15 min. Stained wells were washed 3 times with PBS, and crystal violet was solubilized by the addition of absolute methanol. After incubation for 5 min at room temperature, the solubilized crystal violet was transferred to a new microtiter plate and scanned at 590 nm in a spectrophotometer. A biofilm-forming isolate was defined as one where the average absorbance of the 3 wells was greater than the average absorbance of the negative control wells plus 3 standard deviations (See, e.g., Stepanovic et al., J Microbiol Methods. 2000 April; 40(2):175-9).
  • For biofilm susceptibility testing, isolates shown to produce biofilm by crystal violet testing were grown in triplicate for 48 h as described above. Wells were gently washed twice with PBS before addition of P4075EC, serially diluted as described for planktonic MIC testing. After overnight incubation at 37° C., wells were again washed twice with PBS, and 100 μl of 10% resazurin in MH broth was added to wells. Plates were wrapped in plastic, covered with foil, and again incubated overnight at 37° C. without shaking. Plates were visually inspected the next day, and the minimum biofilm inhibitory concentration (MBIC) was recorded as the lowest concentration of P4075EC in which the wells were a blue color. To calculate minimum biofilm eradication concentrations (MBECs), biofilm was resuspended in the same wells used for MBIC testing by shaking the microtiter plate and then scraping the sides of the wells with a pipet tip. Ten μL were removed from each well, spotted on MH agar plates and incubated at 37° C. After overnight growth, MBECs were recorded as the lowest P4075EC concentration resulting in no growth (See, e.g., Tomlin et al., Can J Microbiol. 2001 October; 47(10):949-54). To confirm initial inoculum concentrations and to quantify viable bacteria in biofilm grown cultures, colonies were enumerated from 10-fold serial dilutions of the 0.5 McFarland inoculating culture and from the untreated positive control wells with resuspended biofilm for MBEC calculations.
  • Sputum preparation. Expectorated sputum, collected from CF patients during the course of routine care, was obtained from the University of Michigan Health System clinical microbiology laboratory and stored at −80° C. Equal volumes of sputum from 15 individuals were pooled, mechanically sheared using a TISSUE MISER homogenizer (FISHER SCIENTIFIC, Pittsburgh, Pa.) at room temperature for 5 min at maximum speed, and then incubated in an 80° C. water bath for 20 min. Processed sputum was divided into 10 ml aliquots, and 100 μl from each aliquot was plated on MH agar and incubated for 48 h at 37° C. to confirm sterility. Aliquots were stored at −80° C.
  • Results.
  • Susceptibility of planktonic bacteria. Due to the opaque white color of P4075EC, the standard CLSI-approved microtiter serial dilution method was modified to include the addition of resazurin as an indicator of bacterial viability. P4075EC was tested in a concentration range of 15.6-2000 μg/ml. MICs were defined as the lowest concentration of P4075EC that did not produce a color change from blue to pink (See FIG. 8). Comparison of this visual inflection point with fluorometric analysis showed that 63% of MIC wells had ≦1% of the metabolic activity of untreated control wells; 91% had ≦5% metabolic activity, and 96% had ≦10% metabolic activity compared to control wells. The MIC results are shown in FIG. 11. All strains were inhibited by the concentrations of P4075EC tested. The MIC50 for the entire panel of 150 strains was 31.2 μg/ml; the MIC90 was 125 μg/ml. Thirty eight strains (25%) were inhibited by the lowest concentration of P4075EC tested (15.6 μg/ml), and only a single strain each required a concentration of 250 μg/ml and 500 μg/ml for inhibition. P4075EC was slightly more active against non-Burkholderia strains (MIC50, 31.2 μg/ml; MIC90 62.5 μg/ml) than Burkholderia strains (MICR, 62.5 μg/ml; MIC 90 125 μg/ml) (See FIG. 9). Activity was comparable across the 10 Burkholderia species tested. No difference was found in the activity of P4075EC against multi-drug resistant Burkholderia strains compared to strains susceptible to one or more antibiotics.
  • To evaluate the bactericidal activity of P4075EC against planktonic bacteria, MBCs were determined on a subset of 34 strains including 22 Burkholderia and 12 non-Burkholderia. All MBCs were within 1 dilution of the respective MICs for this subset of strains except for a single B. cenocepacia strain that had a MIC of 250 μg/ml and an MBC of 2000 μg/ml. A time-kill study of B. multivorans ATCC 17616 showed time- and concentration-dependent killing, with a 99% decrease in bacterial viability within 90 min at a P4075EC concentration two times greater than the MIC and complete killing within 30 min at concentrations eight times greater than the MIC (See FIG. 10A).
  • Susceptibility of bacteria grown in biofilm. To further assess the activity of P4075EC, bacteria grown as biofilms were tested for susceptibility. Crystal violet staining identified 12 biofilm-forming strains from among 25 strains screened from the test panel. Nine (75%) of the 12 strains showed decreased susceptibility to P4075EC, defined as at least a four-fold increase in the MBIC compared to the MIC, when grown as a biofilm (See FIG. 12). The median increase in MBIC compared to the respective MIC of P4075EC for the strains in this set was eight-fold. No evidence of tolerance of biofilm bacteria to P4075EC was observed; the MBEC was the same as the respective MBIC for 10 of the 12 strains.
  • Susceptibility of bacteria in CF sputum. The 12 biofilm-forming strains were also tested for susceptibility to P4075EC in the presence of CF sputum to model more closely the CF pulmonary microenvironment. The MBCs of P4075EC for all 12 strains, grown under planktonic conditions, increased in the presence of 43% sputum (the highest sputum concentration achievable in the microtiter assay) compared to the respective MBCs under standard conditions (See FIG. 12). Nevertheless, all strains remained susceptible to P4075EC in the presence of sputum. CF sputum inhibited the activity of P4075EC against planktonic bacteria in a concentration dependent manner (See FIG. 10B).
  • Example 4 Nanoemulsion Killing of Individual and Mixed Bacterial Biofilms
  • A composition comprising nanoemulsion P4075EC, 7% saline and 20 mM EDTA was tested for its ability to kill bacteria present in biofilms (individual or mixed).
  • Biofilm Growth. Bacterial biofilms on polycarbonate membranes were formed following the procedure described in Current Protocols in Microbiology (John Wiley & Sons, Inc., NJ, USA). Polycarbonate membranes were sterilized by exposing the membrane to UV light for 10 minutes each side using sterile forceps or by steam sterilization at 121° C. for 20 minutes. The sterile membrane was placed on the surface of an agar medium with the shiny surface facing upwards. Overnight culture of the bacterial strain of interest was diluted with broth media to an OD600 of 0.05 and 0.5 μl of the diluted culture placed onto the shiny surface of the polycarbonate membrane on the agar plate medium. The plate with the membrane was incubated at 37° C. for 24 hours following which; the membrane is transferred to a new agar medium for another 24 hours for biofilm formation.
  • Bactericidal Assay for Biofilm Bacteria. Polycarbonate membrane with the biofilm was immersed in 250 μl of 20% P4075EC with 20 mM EDTA and 7% sodium chloride (NaCl) in a 1.5 ml eppendorf tube for 3 hours at 37° C. Seven percent NaCl served as the negative control. Following incubation, the membrane was transferred to 15 ml sterile polypropylene tubes containing 10 ml of sterile PBS. One ml of sterile PBS was added to the eppendorf reaction tubes, vortexed and spun at 3500 rpm for 15 minutes. Following centrifugation, the supernatant containing the NE was removed and the bacterial pellet resuspended in their respective 15 ml tube in which the polycarbonate membrane was collected. The 15 ml tube was then vortexed at maximum speed for 3 minutes to get bacteria from membrane into the PBS. One hundred microliters of undiluted and diluted treated and control samples were plated onto LB agar plates for colony count.
  • As shown in FIGS. 13-16, 20% P4075EC with 7% saline and 20 mM EDTA was effective to kill bacteria present in biofilms comprising a single bacterial strain (See, e.g., FIG. 13, biofilms comprising a plurality of bacterial strains (See, e.g., FIGS. 14-15), as well as biofilms generated from bacteria that escaped killing in a first round of treatment (See, e.g., FIG. 16), attaining between a 4 to 9 log reduction in bacterial numbers.
  • Example 5
  • This example describes the effectiveness of various nanoemulsion formulations against various gram negative pathogens that may be associated with CF, wounds, burns and burn patients, or other environments.
  • As noted above, CF is characterized by chronic respiratory infection that begins early in life with Staphylococcus aureus and Haemophilus influenzae infections and later colonization with mucoid strains of Pseudomonas aeruginosa. In CF lung disease, early colonization with Burkholderia cepacia correlates with a poor prognosis in CF patients. Patients are often prescribed inhaled tobramycin to prevent exacerbations of bacteria infections. With time, patients can become unresponsive to tobramycin therapy/prophylaxis. New inhaled topical agents that are not cross-resistant to known antibiotics would be valued as an alternative to inhaled tobramycin.
  • Several topical nanoemulsions were evaluated for microbiological activity against gram-negative isolates, including P. aeruginosa and B. cepacia. All 3 nanoemulsions kill quickly in the presence of an outer membrane permeabilizer such as EDTA. The nanoemulsions were evaluated against 23 of the 35 gram-negative isolates for cidal activity, and all were bactericidal (85-95%). Thus, the one or more of these novel nanoemulsions can be used to prevent exacerbation of chronic pulmonary infections.
  • Methods: MICs and MBCs were determined using CLSI standard methods. MICs to nanoemulsions were determined in the presence of 5 mM EDTA, a known enhancer of nanoemulsion activity. The addition of alamar blue, a redeox indicator that yields a colorimetric change in response to metabolic activity, was used to determine the MICs of nanoemulsions that are opaque at higher concentrations.
  • Emulsion manufacturing: Nanoemulsions W205EC, P4075EC, and W205GBA2ED are oil-in-water emulsions manufactured from ingredients that are Generally Recognized As Safe (GRAS) with a cationic detergent (cetylpyridinium chloride (CPC) or benzalkonium chloride (BA)) as active ingredients that have proven safe for oral human use. The emulsion is formed from highly purified oil, ethanol, a nonionic surfactant and water. The average nanoemulsion droplet size was 180 nm for W205EC and 350 nm for P4075EC and W205GBA2ED, as measured by dynamic light scattering using a Malvern Zetasizer Nano3600 (Malvern Instruments Ltd., Worcestershire, UK). The formulations for W205EC and P4075EC are described above. W205GBA2ED (v/v %) is distilled water (18.93%), TWEEN 20 (5%), Glycerine (8%), Soybean oil (64%), BTC 824 50% (4%) (Stepan, Northfield, Ill.), and EDTA (0.0745%). The formulation may be diluted in water (e.g., 60% W205GBA2ED, 40% water) to provide 60% W205GBA2ED. This material may be diluted to a 10% formulation with 10 mM EDTA (e.g., 76 g water per 24 g 100 mM EDTA per 20 g 60% W205GBA2ED). Thus, in some embodiments, a formulation comprising oil, water, glycerine, surfactant, and BTC (n-alkyl dimethyl benzyl ammonium chloride), and/or EDTA is provided.
  • Source of isolates. The source of the clinical isolates were blood stream or skin and soft tissue isolates collected by JMI Laboratories in the past 2 years.
  • MIC and MBC determinations. MICs of W205EC±0, 5, 10, 15, and 20 mM EDTA were evaluated in cation-adjusted Mueller-Hinton broth by microdilution per M7-A7 (2006). MICs for a panel of gram-negative isolates were determined for the nanoemulsions, W205EC, P4075EC, and W205GBA2ED in the presence of 5 mM EDTA. EDTA was used to permeabilize the gram-negative envelope, aiding infusion of the nanodroplets to the cell membrane, resulting in lysis. Alamar blue was added to the assay panels 2 hours post-inoculation for enhanced MIC endpoint detection. MBC values were assessed for all nanoemulsions and a comparator compound (fusidicacid) by plating the entire broth content from the MIC well and from those four doubling dilutions above the MIC for each selected organism onto blood agar growth media. Quantitative colony counts were performed on the initial inoculum. The lowest concentration of antimicrobial agent that killed≧9.9% of the starting test inoculum was defined as the MBC endpoint. See Tables 2, 3, and 4.
  • TABLE 2
    MIC values of W205EC + EDTA
    Bacterial
    Isolates
    0 mM 5 mM 10 mM 15 mM 20 mM
    K. pneumoniae 64  8-16 8 8 8
    24-5A
    E. coli ATCC 16 1 ≦0.12 ≦0.12 ≦0.12
    25922
    P. mirabilis 128 ≦0.12* ≦0.12 ≦0.12 ≦0.12
    119-163A
    A. baumannii 32 ≦0.12* ≦0.12 ≦0.12 ≦0.12
    67-299A
    P. aeruginosa >256 32-64 32 32 32
    ATCC 27853
    B. cepacia >256 >256 >256 >256 >256
    30-492A
    *EDTA alone inhibited these strains.
  • TABLE 3
    Cidality of nanoemulsions against a subset (23)
    of gram-negative isolates
    MBC/
    MIC Compound (number of strains)
    ratio W205EC P4075EC W205GBA2ED Levofloxacin
    Cidal ↑ 1 7 10 9 9
    2 6 7 7 8
    4 4 2 2 1
    Static ↓ 8 1 1 1
    >8x 2 1 5
    No 3 3 3
    growth*
  • TABLE 4
    Escherichia coli (n = 5)a Klebsiella pneumoniae (n = 5)
    % %
    Antimicrobial susceptible/ susceptible/
    agent MIC50 MIC90 Range resistantb MIC50 MIC90 Range resistant
    W205EC 2 1-2 —/— 8  4-16 —/—
    P4075EC 4 4 —/— 8  8-16 —/—
    W205G BA2 ED 2 2-4 —/— 4 4-8 —/—
    Ceftazidime <=1 <=1->16 80.0/20.0 <=1 <=1->16 80.0/20.0
    Cefepime <=0.12 <=0.12->16   80.0/20.0 <=0.12 <=0.12->16   80.0/20.0
    Piperacillin/ 2 1-8 100.0/0.0  2  1-64 80.0/0.0 
    tazobactam
    Imipenem 0.25 <=0.12-0.5   100.0/0.0  0.25 <=0.12-0.25  100.0/0.0 
    Gentamicin <=2 <=2->8  80.0/20.0 <=2 <=2 100.0/0.0 
    Tobramycin 0.5 0.25-16   80.0/20.0 0.25 0.25-16   80.0/20.0
    Levofloxacin 0.03 0.03->8   60.0/40.0 0.06 0.06-8   80.0/20.0
    Tetracycline >8  4->8 20.0/80.0 <=2 <=2-4  100.0/0.0 
    Colistin <=0.5 <=0.5 —/— <=0.5 <=0.5 —/—
    Proteus mirabilis (n = 5) Pseudomonas aeruginosa (n = 5)
    % %
    Antimicrobial susceptible/ susceptible/
    agent MIC50 MIC90 Range resistant MIC50 MIC90 Range resistant
    W205EC 8 4-8 —/— 16 16-32 —/—
    P4075EC 16  8-16 —/— 64 32-64 —/—
    W205G BA2 ED 8  4-16 —/— 16  8-16 —/—
    Ceftazidime <=1 <=1 100.0/0.0  2 2-4 100.0/0.0 
    Cefepime <=0.12 <=0.12 100.0/0.0  4 1-8 100.0/0.0 
    Piperacillin/ <=0.5 <=0.5-1    100.0/0.0  2  2-16 100.0/0.0 
    tazobactam
    Imipenem 1 <=0.12-2    100.0/0.0  1  1->8 80.0/20.0
    Gentamicin <=2 <=2 100.0/0.0  <=2 <=2-4  100.0/0.0 
    Tobramycin 1 0.25-1   100.0/0.0  0.5 0.25-1   100.0/0.0 
    Levofloxacin 0.12 0.06-2   100.0/0.0  1 0.25-4   80.0/0.0 
    Tetracycline >8 >8  0.0/100.0 8  8->8  0.0/40.0
    Colistin >4 >4 —/— 1 <=0.5-2    100.0/0.0 
    Acinetobacter baumannii (n = 5)b Stenotrophomonas maltophilia (n = 5)c
    % %
    Antimicrobial susceptible/ susceptible/
    agent MIC50 MIC90 Range resistant MIC50 MIC90 Range resistant
    W205EC <=0.12 <=0.12-2    —/— <=0.12 <=0.12 —/—
    P4075EC <=0.12 <=0.12-4    —/— <=0.12 <=0.12 —/—
    W205G BA2 ED <=0.12 <=0.12-4    —/— <=0.12 <=0.12 —/—
    Ceftazidime 4 <=1->16 80.0/20.0 16  2->16 40.0/40.0
    Cefepime 2 <=0.12->16   80.0/20.0 >16  4->16 —/—
    Piperacillin/ 2 <=0.5->64   80.0/20.0 >64  8->64 —/—
    tazobactam
    Imipenem <=0.12 <=0.12->8    80.0/20.0 >8  4->8 —/—
    Gentamicin <=2 <=2->8  80.0/20.0 >8 <=2->8  —/—
    Tobramycin 0.5 0.25->16  80.0/20.0 >16  0.5->16 —/—
    Levofloxacin 0.25 0.06->8   80.0/20.0 0.5 0.5-8   60.0/20.0
    Tetracycline <=2 <=2->8  80.0/20.0 >8 <=2->8  —/—
    Colistin <=0.5 <=0.5-2    100.0/0.0  <=0.5 <=0.5-1    —/—
    Burkholderia cepacia (n = 5)
    %
    Antimicrobial susceptible/
    agent MIC50 MIC90 Range resistant
    W205EC 256  64->256 —/—
    P4075EC 256  128->256 —/—
    W205G BA2 ED 128  32->256 —/—
    Ceftazidime 2 2-4 100.0/0.0 
    Cefepime 8  4-16 —/—
    Piperacillin/ 4  2-32 —/—
    tazobactam
    Imipenem 8 4-8 —/—
    Gentamicin >8 >8 —/—
    Tobramycin >16 >16  —/—
    Levofloxacin 2 1-2 100.0/0.0 
    Tetracycline >8 >8 —/—
    Colistin >4 —/—
  • Results: All 3 nanoemulsions had activity against Gram-negative isolates (EDTA alone inhibited 3 and 5 strains of A. baumannii and S. maltophilia, respectively). This included isolates that were resistant to comparator agents. MBCs were performed for 23 isolates; W205EC, P4075EC, and W205GBA2ED were bactericidal against 85, 95, and 90% of the isolates, respectively. No cross-resistance to any known antibiotic was observed for any of the nanoemulsions.
  • Conclusions: Nanoemulsions were broadly active against Gram-negative species, including multidrug-resistant isolates. The documented MICs were within the range of concentrations achievable with topical application to skin or mucosal tissues. One or more of the nanoemulsions may be useful for prophylaxis and/or therapeutic treatment of chronic pulmonary infections in cystic fibrosis patients.
  • Example 6
  • The purpose of this example was to determine if nanoemulsions according to the invention have activity against Haemophilus influenzae isolates. H. influenzae is an important respiratory pathogen implicated in acute exacerbations of cystic fibrosis patients as well as upper and lower respiratory tract infections for non-CF normal individuals.
  • This example determined the minimum inhibitory concentration of three exemplary nanoemulsions (W205EC, P4075EC and W205GBA2) plus comparator drugs against clinical isolates of H. influenzae. The results, as described in more detail below, show that P4075EC and W205EC had similar MIC ranges of 1.88-3.75 μg/ml. W205GBA2 had a MIC range of 8-16 μg/ml. The addition of EDTA at 1 mM to each well containing nanoemulsion had no effect on the MIC. Concentrations of EDTA greater than 1 mM inhibited the growth of bacteria, likely reducing the concentration of cations to levels below those that support growth. MBC data for the three nanoemulsions+1 mM EDTA were obtained for seven strains. The MBCs were within 4-fold of the respective MICs, consistent with each nanoemulsion having bactericidal activity against H. influenzae isolates.
  • A. Materials and Methods
  • Source of drugs. W205EC, lot x1151, at a concentration of 5000 μg/ml (stock concentration) was a pooled sample of clinical trial material (NB-001), manufactured at Patheon, Mississauga, Ontario, Canada. P4075EC, lot X1138, and W205GBA2, lot X1103, were manufactured at NanoBio Corporation. Comparator compounds, azithromycin, ampicillin, cefepime and clindamycin were purchased from The United States Pharmacopeia; catalog numbers were 1046056, 1033000, 1097636, 1136002, respectively. Tetracycline was purchased from Fluka Biochemika, catalog number 87128. The emulsions were produced by mixing a water immiscible oil phase with an aqueous phase. The base formulations are shown in Table 5 below and represent the neat emulsions, which were further diluted to the desired %. Compositions are w/w % unless otherwise noted.
  • TABLE 5
    Nanoemulsions
    Nanoemulsion Component Weight Percent (w/w %)
    W205EC ED Distilled Water 23.418% 
    EDTA 0.0745% 
    Cetylpyridinium Chloride 1.068%
    Tween
    20  5.92%
    Ethanol  6.73%
    Soybean Oil 62.79%
    P4075EC Distilled Water 23.49%
    CPC 1.068%
    Poloxamer
    407  5.92%
    Ethanol  6.73%
    Soybean Oil, NP 62.79%
    W205GBA2 (v/v %) Distilled Water 20.93%
    BTC 824    2%
    Tween
    20    5%
    Glycerine
       8%
    Soybean Oil   64%
  • Source of bacterial strains. Ten clinical isolates of H. influenzae were obtained from Case Western Reserve University, Cleveland, Ohio. The quality control isolate, ATCC 49247, was obtained from the American Type Culture Collection.
  • Susceptibility assays. The susceptibility of H. influenzae isolates to five commercial antibiotics and three nanoemulsion formulations was evaluated using the guidelines published by Clinical Laboratory Standards Institute (Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility and Testing; Seventeenth Informational Supplement. CLSI document MI00-S17 (ISBN 1-56238-625-5). CLSI, 940 West Valley Road, Suite 1400, Wayne, Pa. 19087-1988, 2007). The ranges of drug concentrations tested were as follows: Azithromycin at 0.25-128 μg/ml, Ampicillin at 0.063-32 μg/ml, Cefepime at 0.063-32 μg/ml, Clindamycin at 0.125-64 μg/ml, Tetracycline at 0.125-64 μg/ml, P4075EC at 0.12-60 μg/ml cetylpyridinium chloride, W205EC at 0.12-60 μg/ml cetylpyridinium chloride, W205GBA2 at 0.125-64 μg/ml benzalkonium chloride. Since nanoemulsions are not a single component, the MICs/MBCs are expressed as the concentration of the cationic surfactant; W205EC and P4075EC both contain cetylpyridinium chloride (CPC) as the cationic surfactant, while W205GBA2 contains benzalkonium chloride (BA) as the cationic surfactant. Isolates were also tested with the same range of nanoemulsion concentrations+1-5 mM EDTA to see if the latter enhanced the activity of the nanoemulsion. EDTA concentrations were tested alone to ensure that chelation of cations by EDTA was sufficient to inhibit bacterial growth independent of the nanoemulsion.
  • Minimum inhibitory concentrations (MIC) were determined visually as the first totally clear well in the series of the 10 concentrations of each drug. For the nanoemulsions, because they are intrinsically opaque, the MIC was determined as the first well observed to have some clearing from the least concentrated to the most concentrated of the serial dilution. MBC values were assessed for the nanoemulsions+1 mM EDTA for 7 clinical strains and the ATCC 49257 isolate by plating 10 μl from the well determined to be the MIC and 4 wells above the MIC onto chocolate blood agar media. The lowest concentration of antimicrobial agent that killed≧99.9% of the starting test inoculum was defined as the MBC. A compound or nanoemulsion is defined as bactericidal if its MBC/MIC ratio was ≦4.
  • Results: Data for the MICs and MBCs are shown in Tables 6-7. P4075EC, W205EC, and W205GBA2 were equally effective antimicrobials in the presence or absence of 1 mM EDTA. The MIC90 values for these compounds were 3.75, 3.75, and 16 μg/ml, respectively (Table 6). By MIC90 values, the majority of isolates were susceptible to azithromycin, cefepime, tetracycline and ampicillin, but were resistant to clindamycin and ampicillin.
  • The MBC values were within 4-fold of the respective MICs for each isolate tested (Table 7); thus each of the nanoemulsions were bactericidal for H. influenzae, with a range of MBCs for P4075EC, W205EC, and W205GBA2 of 1.88-7.5, 1.88-7.5 and 8-16 μg/ml, respectively.
  • Conclusions: P4075EC and W205EC appeared equally effective against clinical isolates of H. influenzae. The benzalkonium chloride formulation, W205GBA2, was two- to four-fold less active than the other nanoemulsions by MIC90 and MBC values. EDTA at concentrations≧2 mM EDTA inhibited the growth of H. influenzae under these growth conditions. The addition of EDTA was not necessary to enhance the activity of any of the nanoemulsions.
  • TABLE 6
    Susceptibility of H. influenzae clinical isolates to nanoemulsions and control
    antibiotics
    MIC (μg/ml)
    P4075EC + W205EC + W205GBA2 +
    1 mM 1 mM 1 mM
    Strain P4075ECa EDTA W205ECa EDTA W205GBA2 b EDTA Azic Ampc Cefc Tetc Clic
    49247   1.88 1.88 1.88 0.94 8 4 1 4 1 16 8
    30 1.88 0.94 1.88 0.94 4 8 2 0.25 0.125 0.5 16
    32 1.88 1.88 1.88 3.75 8 4 8 32 0.125 2 8
    33 3.75 1.88 3.75 3.75 8 8 1 0.25 0.0625 1 16
    34 3.75 3.75 3.75 3.75 8 8 1 >32 0.125 1 16
    35 3.75 3.75 7.5 7.5 16 16 1 0.25 0.125 0.5 8
    36 3.75 3.75 3.75 3.75 8 16 2 0.125 0.125 1 16
    37 1.88 1.88 3.75 3.75 16 16 1 >32 0.125 1 4
    38 3.75 3.75 3.75 3.75 16 16 1 0.25 0.0625 0.5 16
    41 1.88 0.94 1.88 0.94 8 4 2 >32 0.25 1 8
    42 1.88 1.88 1.88 3.75 8 8 1 >32 0.125 1 4
    MIC 1.88-3.75 0.94-3.75 1.88-3.75 0.94-7.5 4-16 4-16 1-8 0.25->32 0.0625-1 0.5-16 4-16
    range
    MIC90 3.75 3.75 3.75 3.75 16 16 2 >32 0.125 2 16
    aMIC value reflects the amount of μg cetylpyridinium chloride/ml
    bMIC values reflects the amount of μg benzalkonium chloride/ml
    cAzi = azithromycin; Amp = ampicillin; Cef = cefepime; Tet = tetracycline; Cli = clindamycin
  • TABLE 7
    Cidality of nanoemulsions against H. influenzae clinical isolate
    MBC (μg/ml)
    P407 5EC + W205EC + 1 mM W205GBA2 +
    Strain 1 mM EDTA EDTA 1 mM EDTA
    49247 1.88 1.88 8
    30 1.88 1.88 8
    32 3.75 3.75 8
    34 7.5 3.75 8
    35 7.5 7.5 16
    36 7.5 7.5 16
    37 3.75 3.75 8
    38 7.5 7.5 16
  • For Table 8 below, drug concentrations are in μg/ml for the comparators, as μg cetylpyridinium chloride (CPC)/ml for W205EC and P4075EC and as μg benzalkonium chloride (BA)/ml for W205GBA2. NC is for Negative Control and GC is for Growth Control. The chart describes the wells of a 96 well microtiter plate. The rows listed as I, J, K, and L are the rows of a partial plate used to evaluate the nanoemulsions with different EDTA concentrations. For each clinical strain and control strain, the chart below was used for tracking the MIC and MBC data. If the wells were too opaque to read the MIC ( ), the MBC was relied upon to provide the MIC. In Table 8, contain plate counts for MBC. The second number is the plate count of a 10 μl sample from the microtiter plate used to determine the MBC. The MBC was defined as ≧3-log reduction of the inoculum. The darker highlighting corresponds to MIC.
  • TABLE 8
    H. influenzae MIC of Nanoemulsions with EDTA
    Figure US20100092526A1-20100415-C00001
    Figure US20100092526A1-20100415-C00002
    Figure US20100092526A1-20100415-C00003
    Figure US20100092526A1-20100415-C00004
    Figure US20100092526A1-20100415-C00005
    Figure US20100092526A1-20100415-C00006
    Figure US20100092526A1-20100415-C00007
    Figure US20100092526A1-20100415-C00008
    Figure US20100092526A1-20100415-C00009
    Figure US20100092526A1-20100415-C00010
    Figure US20100092526A1-20100415-C00011
    Highlighted in Green are rows containing plate counts for the MBC. The second number is the plate count of a 10 μl sample from the microtiter plate used to determine the MBC. The MBC was defined as ≧3-log reduction of the inoculum.
    Azi = azithromycin:
    Amp = ampicillin:
    Cef = cefepime:
    Cli = clindamycin:
    Tet = tetracycline.
  • Example 7
  • The purpose of this study was to determine the MIC and MBC of four nanoemulsions (NEs) according to the invention and compare the performance of the NEs to conventional antibiotics against MRSA. MRSA is one of the microorganisms that cause lung infections in young cystic fibrosis patients and is also implicated in infections of skin and soft tissue, including burn wound infections. Because of its varied virulence mechanisms, infections caused by MRSA can be lethal.
  • The purpose of this example was to evaluate 4 nanoemulsions according to the invention for use in the topical treatment of infectious diseases. The nanoemulsions tested were W805EC, W205ECEDL2, W205GBA2ED and P4075EC. This example determined the minimum inhibitory concentration (MIC) of the four nanoemulsions plus comparator drugs against 29 clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA).
  • The results shown below demonstrate that the MIC ranges for the nanoemulsions were 0.5-2 (W805EC), 0.5-4 (W205ECEDL2), 1-4 (W205GBA2ED), 1-4 μg/ml (P4075EC), respectively. Minimum bactericidal concentration (MBC) data was also collected and the MBC ranges were 0.5-16 (W805EC), 1->32 (W205ECEDL2), 2-16 (W205GBA2ED), and 1->16 μg/ml (P4075EC), respectively.
  • A. Methods and Materials
  • Source of drugs and isolates. Twelve comparator compounds, clindamycin (USP #1136002), doxycycline (USP #1226003), erythromycin (Sigma #E0774), levofloxacin (Sigma #28266), mafenide acetate (USP #1373008), mupirocin (Sigma #M7694), silvadene (USP #61260), vancomycin (Sigma #V1764), Oxacillin (Sigma #28221), sulfamethoxazole (USP #1631001), trimethoprim (USP # 1692505), and silver nitrate (VWR #VW3462-0) were compared to the four nanoemulsions. The four nanoemulsions were (a) W805EC, (b) W205ECEDL2, (c) W205GBA2ED and (d) P4075EC. See Table 10, which provides drug panel templates (with increasing dilutions across the panel). The emulsions were produced by mixing a water immiscible oil phase with an aqueous phase. The W205ECEDL2 formulation was microfluidized. The base formulations are shown in Table 9 below and represent the neat emulsions, which were further diluted to the desired %. Compositions are w/w % unless otherwise noted.
  • TABLE 9
    Nanoemulsions
    Weight Percent
    Nanoemulsion Component (w/w %)
    W805EC Water 23.490% 
    Ethanol 6.730%
    Cetylpyridinium Chloride 1.068%
    Polysorbate
    80 5.920%
    Refined Soybean Oil 62.790% 
    W205ECEDL2 Distilled Water 23.418% 
    EDTA 0.0745% 
    Cetylpyridinium Chloride 1.068%
    Tween
    20  5.92%
    Ethanol  6.73%
    Soybean Oil 62.79%
    W205GBA2ED (v/v %) Distilled Water 20.93%
    EDTA 0.0745% 
    BTC 824    2%
    Tween
    20    5%
    Glycerine
       8%
    Soybean Oil   64%
    P4075EC Distilled Water 23.49%
    CPC 1.068%
    Poloxamer
    407  5.92%
    Ethanol  6.73%
    Soybean Oil, NP 62.79%
  • TABLE 10
    Drug Panel Templates
    Drug Panel Templates
    Compound in μg/ml (% for AgNO3)
    Compound 1 2 3 4 5 6 7 8 9 10 11 12
    A Clindamycin 64 32 16 8 4 2 1 0.5 0.25 0.125 NC GC
    B Doxycycline 128 64 32 16 8 4 2 1 0.5 0.25 NC GC
    C Erythromycin 128 64 32 16 8 4 2 1 0.5 0.25 NC GC
    D Levofloxacin 64 32 16 8 4 2 1 0.5 0.25 0.125 NC GC
    E Mafenide acetate 128 64 32 16 8 4 2 1 0.5 0.25 NC GC
    F Mupirocin 64 32 16 8 4 2 1 0.5 0.25 0.125 NC GC
    G Silvadene 128 64 32 16 8 4 2 1 0.5 0.25 NC GC
    H Vancomycin 64 32 16 8 4 2 1 0.5 0.25 0.125 NC GC
    A Oxacillin 16 8 4 2 1 0.5 0.25 0.125 0.0625 0.03125 NC GC
    B Sulfamethazole 32 16 8 4 2 1 0.5 0.25 0.125 0.0625 NC GC
    C Trimethoprim 64 32 16 8 4 2 1 0.5 0.25 0.125 NC GC
    D AgNO3 0.01 0.005 0.0025 0.00125 0.00063 0.00031 0.00016 0.00008 0.00004 0.00002 NC GC
    E P4075EC 32 16 8 4 2 1 0.5 0.25 0.125 0.0625 NC GC
    F W205ECEDL2 32 16 8 4 2 1 0.5 0.25 0.125 0.0625 NC GC
    G W205GBA2ED 64 32 16 8 4 2 1 0.5 0.25 0.125 NC GC
    H W805EC 32 16 8 4 2 1 0.5 0.25 0.125 0.0625 NC GC
  • Source of bacterial strains. Twenty-nine clinical isolates of MRSA were obtained from the University of Dentistry and Medicine of New Jersey. The quality control isolate, ATCC 29213, was obtained from the American Type Culture Collection. Table 11 below provides the phenotype/genotype of each strain.
  • TABLE 11
    Phenotype and genotype of MRSA isolates
    Strain Phenotype or Genotype
    Number MLST spatype spa repeat pattern SCCmec PFGE PVL
     2394 ST59 17 ZI-DI-MI-DI-MI-NI-KI-BI IV USA1000 +
     2402
     2926 ST5 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
     9897 ST1 131 UI-JI-JI-FI-KI-BI-PI-EI IV USA400 +
    11118 131 UI-JI-JI-FI-KI-BI-PI-EI IV USA400 +
    11512 ST36 16 WI-GI-KI-AI-KI-AI-OI-MI-QI-QI-QI II
    11540 ST8 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV USA300 +
    11554 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV +
    13219 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI IV
    13367 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    13386 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    13408 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI IV
    13606 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    13610 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI I
    13643 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV +
    13693 7 YI-HI-GI-CI-MI-BI-QI-BI-LI-OI IV
    13701 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    13722 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    13756 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    13759 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV
    13868 2 TI-JI-MI-BI-MI-DI-MI-GI-MI-KI II
    15337 18 WI-FI-KI-AI-OI-MI-QI II
    18998 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV USA300 +
    (MUP-R)
    19001 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV USA300 +
    (MUP-R)
    19017 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV USA300 +
    (MUP-R)
    19024 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV USA300 +
    (MUP-R)
    19047 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV +
    (MUP-R)
    19069 ST8 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV USA300 +
    (MUP-R)
    19156 1 YI-HI-GI-FI-MI-BI-QI-BI-LI-OI IV +
    (MUP-R)
  • Susceptibility assays. The susceptibility of MRSA to twelve traditional antimicrobials and four nanoemulsion formulations was tested using the guidelines published by the Clinical Laboratory Standards Institute (Clinical and Laboratory Standard Institute, “Antimicrobial Susceptibility and Testing; Seventeenth Informational Supplement. CLSI document M100-S17 (ISBN 1-56238-625-5), CLSI, Wayne, Pa.). The ranges of drug concentrations were as follows: clindamycin (64-0.125 μg/ml), doxycycline (128-0.25 μg/ml), erythromycin (128-0.25 μg/ml), levofloxacin (64-0.125 μg/ml), mafenide acetate (128-0.25 μg/ml), mupirocin (64-0.125 μg/ml), silvadene (128-0.25 μg/ml), vancomycin (64-0.125 μg/m), oxacillin (16-0.03125 μg/ml), sulfamethoxazole (32-0.0625 μg/ml), trimethoprim (64-125 μg/ml), silver nitrate (0.01-0.00002%), W805EC (32-0.0625 μg/ml), W205ECEDL2 (32-0.0625 μg/ml), W205GBA2ED (64-0.125 μg/ml), and P4075EC (32-0.0625 μg/ml).
  • Minimum inhibitory concentrations (MIC) were determined visually as the first totally clear well in the series of the 10 concentrations of each drug. For the nanoemulsions, because they are intrinsically opaque, the MIC was called as the first well observed to have some clearing from the least concentrated to the most concentrated. Minimum bactericidal concentrations (MBC) values were assessed for all the antimicrobials tested. The lowest concentration of antimicrobial agent that killed 2: 99.9% of the starting test inoculum was defined as the MBC. A compound or nanoemulsion is defined as bactericidal if its MBC/MIC ratio was ˜4.
  • B. Results
  • Data for the MICs and MBCs are shown in Tables 11 and 12. All four nanoemulsions were potent against the clinical isolates of MRSA, with no differential activity noted for community-acquired MRSA (SCCmec type IV) or hospital-associated MRSA (SCCmec types I-III). Further, there appeared to be no cross-resistance of the nanoemulsions to any of the known antibiotics. The MIC90 values were 2 μg/ml for P4075EC, W205ECEDL2 and W805EC. W205GBA2ED had a MIC90 Of 4 μg/ml. The MIC90 values indicated that this collection of MRSA was susceptible to oral antibiotics, doxycyline (MIC90=4 μg/ml), sulfamethoxazole (MIC90=4 μg/ml), trimethoprim (MIC90=0.5 μg/ml), and vancomycin (MIC90=1 μg/ml). Isolates were generally resistant to oral antibiotics levofloxacin (MIC90=32 μg/ml), clindamycin (MIC90=>64 μg/ml) and erythromycin (MIC90=>128 μg/ml). Topical antibiotics that are used in the treatment of burn wound infections, silvadene and silver nitrate, had MIC90 values of 32 μg/ml and 0.00063%, respectively. Mafenide acetate (Sulfamylon) is another topical treatment for burn wound infections, but it was uniformly inactive against MRSA (MIC90=>128 μg/ml). Mupirocin is a topical treatment for skin and soft tissue infections and is also used to eradicate carriage of MRSA; 31% of the isolates were resistant to this antibiotic.
  • The MBC90 values for all of the nanoemulsions were within four-fold of the respective MIC90 values, indicating that the nanoemulsions are bactericidal to ˜90% of the isolates. Among the other 12 antibiotics, only vancomycin was bactericidal.
  • TABLE 12
    Susceptibility of MRSA to nanoemulsions and comparator compounds: MIC
    analysis
    Figure US20100092526A1-20100415-P00899
    MIC (μg/ml)
    Rank Maf.
    Order Clind Dox Eryth Levo Acet Mup Silva Vanco Ox Sulfa
    29 >64 4 >128 >64 >128 >64 64 2 >16 >32
    28 >64 4 >128 >64 >128 >64 64 1 >16 >32
    27 >64 4 >128 32 >128 >64 32 1 >16 4
    26 >64 4 >128 32 >128 >64 32 1 >16 4
    25 >64 0.25 >128 32 >128 >64 32 1 >16 2
    24 >64 0.25 >128 32 >128 >64 32 1 >16 2
    23 >64 0.25 >128 32 >128 >64 32 1 >16 2
    22 >64 0.25 >128 16 >128 >64 32 1 >16 1
    21 >64 0.25 >128 16 >128 16 32 1 >16 1
    20 >64 0.25 >128 16 >128 0.25 32 1 >16 1
    19 >64 0.25 >128 16 >128 0.25 32 1 >16 1
    18 >64 0.25 >128 8 >128 0.25 32 1 >16 1
    17 >64 0.25 >128 8 >128 0.25 32 1 >16 1
    16 >64 0.25 128 8 >128 0.25 32 1 >16 1
    15 >64 0.25 64 8 >128 0.25 32 1 >16 1
    14 >64 0.25 64 8 >128 0.25 32 1 >16 1
    13 >64 0.25 64 4 >128 0.25 32 1 >16 1
    12 0.125 0.25 64 4 >128 0.25 32 1 >16 0.5
    11 0.125 0.25 64 4 >128 0.125 32 1 >16 0.5
    10 0.125 0.25 64 4 >128 0.125 32 1 >16 0.5
     9 0.125 0.25 32 4 >128 0.125 32 1 >16 0.5
     8 0.125 0.25 32 2 >128 0.125 32 0.5 >16 0.5
     7 0.125 0.25 32 0.5 >128 0.125 32 0.5 >16 0.5
     6 0.125 0.25 16 0.25 >128 0.125 16 0.5 >16 0.25
     5 0.125 0.25 8 0.25 >128 0.125 8 0.5 >16 0.25
     4 0.125 0.25 0.25 0.25 >128 0.125 8 0.5 >16 0.25
     3 0.125 0.25 0.25 0.125 >128 0.125 8 0.5 16 0.25
     2 0.125 0.25 0.25 0.125 >128 0.125 8 0.5 16 0.25
     1 0.125 0.25 0.25 0.125 128 0.125 8 0.5 0.5 0.0625
    MIC50(15) >64 0.25 64 8 >128 0.25 32 1 >16 1
    MIC90(26) >64 4 >128 32 >128 >64 32 1 >16 4
    MIC (μg/ml)
    Rank Silver W205EC W205G
    Order Trim Nitrate P4075EC ED L2 BA2 ED W805EC
    29 >64 0.00250 4 4 4 2
    28 1 0.00125 4 2 4 2
    27 1 0.00125 2 2 4 2
    26 0.5 0.00626 2 2 4 2
    25 0.25 0.00626 2 2 4 2
    24 0.125 0.00626 2 2 4 2
    23 0.125 0.00626 2 2 2 2
    22 0.125 0.00626 2 2 2 1
    21 0.125 0.00626 2 2 2 1
    20 0.125 0.00626 2 2 2 1
    19 0.125 0.00312 2 1 2 1
    18 0.125 0.00312 2 1 2 1
    17 0.125 0.00312 1 1 2 1
    16 0.125 0.00312 1 1 2 1
    15 0.125 0.00312 1 1 2 1
    14 0.125 0.00312 1 1 2 1
    13 0.125 0.00312 1 1 2 1
    12 0.125 0.00312 1 1 2 1
    11 0.125 0.00312 1 1 2 1
    10 0.125 0.00312 1 1 2 1
     9 0.125 0.00312 1 1 2 1
     8 0.125 0.00312 1 1 2 1
     7 0.125 0.00312 1 1 2 1
     6 0.125 0.00312 1 1 2 1
     5 0.125 0.00312 1 1 2 1
     4 0.125 0.00312 1 1 2 1
     3 0.125 0.00312 1 1 1 0.5
     2 0.125 0.00312 1 0.5 1 0.5
     1 0.125 0.00312 1 0.5 1 0.5
    MIC50(15) 0.125 0.00031 1 1 2 1
    MIC90(26) 0.5 0.00063 2 2 4 2
    Figure US20100092526A1-20100415-P00899
    indicates data missing or illegible when filed
  • TABLE 13
    Susceptibility of MRSA to nanoemulsions and comparator compounds: MBC
    analysis
    Figure US20100092526A1-20100415-P00899
    MBC (μg/ml)
    Maf.
    Rank Order Clind Dox Eryth Levo Acet. Mup Silva. Vanco Ox Sulfa
    29 >64 >128 >128 >64 >128 >64 >128 4 >16 >32
    28 >64 >64 >128 >64 >128 >64 >128 4 >16 >32
    27 >64 >64 >128 >64 >128 >64 >128 4 >16 >32
    26 >64 >64 >128 >64 >128 >64 >128 2 >16 >32
    25 >64 >64 >128 >64 >128 >64 >128 2 >16 >32
    24 >64 >4 >128 >64 >128 >64 >128 2 >16 >32
    23 >64 >4 >128 64 >128 >64 >128 2 >16 >32
    22 >64 >4 >128 64 >128 >64 >128 2 >16 >16
    21 >64 >4 >128 >32 >128 >64 >128 2 >16 >16
    20 >64 >4 >128 32 >128 >8 >128 2 >16 >16
    19 >64 >4 >128 32 >128 >4 >128 1 >16 >16
    18 >64 >4 >128 32 >128 >4 >128 1 >16 >16
    17 >64 >4 >128 32 >128 >4 >128 1 >16 >16
    16 >64 >4 >128 32 >128 >4 >128 1 >16 >16
    15 >64 >4 >128 16 >128 >4 >128 1 >16 >16
    14 >64 >4 >128 16 >128 >2 128 1 >16 >16
    13 >64 >4 >128 16 >128 >2 128 1 >16 >16
    12 >2 >4 >128 8 >128 >2 128 1 >16 >8
    11 >2 >4 >128 8 >128 >2 128 1 >16 >8
    10 >2 >4 >128 8 >128 >2 128 1 >16 >8
     9 >2 >4 >128 8 >128 >2 128 1 >16 >8
     8 >2 >4 >128 4 >128 >2 128 1 >16 >8
     7 >2 >4 >128 1 >128 >2 128 1 >16 >4
     6 >2 >4 >128 0.5 >128 >2 64 1 >16 >4
     5 >2 >4 64 0.25 >128 >2 64 1 >16 >4
     4 >2 >4 >4 0.25 >128 >2 64 1 >16 >4
     3 >2 2 >4 0.25 >128 1 64 0.5 >16 >1
     2 2 1 >4 0.25 >128 1 64 0.5 >16 1
     1 1 1 >4 0.25 >128 1 64 0.5 1 0.0625
    MBC50(15) >64 >4 >128 16 >128 >2 128 1 >16 >16
    MBC90(26) >64 >64 >128 >64 >128 >64 >128 2 >16 >32
    MBC (μg/ml)
    Silver W205EC W205G
    Rank Order Trim Nitrate P4075EC ED L2 BA2 ED W805EC
    29 >64 0.01 >16 >32 16 16
    28 >16 >0.01 16 8 8 4
    27 >16 >0.01 16 8 8 4
    26 >4 >0.01 8 8 8 4
    25 >2 >0.005 8 8 8 4
    24 >2 >0.005 8 4 8 4
    23 >2 >0.005 4 4 4 4
    22 4 0.005 4 4 4 4
    21 2 0.0025 4 4 4 4
    20 2 0.0025 4 4 4 4
    19 2 0.0025 4 4 4 4
    18 1 0.0025 4 4 4 2
    17 1 0.0025 4 4 4 2
    16 0.5 0.0025 4 4 4 2
    15 0.5 0.0025 2 4 4 2
    14 0.5 0.0025 2 2 4 2
    13 0.5 0.0025 2 2 4 2
    12 0.5 0.0025 2 2 4 2
    11 0.25 0.0025 2 2 4 2
    10 0.25 0.0025 2 2 4 2
     9 0.25 0.00125 2 2 4 2
     8 0.25 0.00125 2 2 4 2
     7 0.25 0.00125 2 2 4 2
     6 0.25 0.00125 2 2 4 2
     5 0.125 0.00125 2 2 4 1
     4 0.125 0.00125 2 2 4 1
     3 0.125 0.00125 2 2 2 1
     2 0.125 0.00125 1 1 2 1
     1 0.0125 0.00125 1 1 2 0.5
    MBC50(15) 0.5 0.0025 2 2 4 2
    MBC90(26) >4 >0.01 8 8 8 4
    Figure US20100092526A1-20100415-P00899
    indicates data missing or illegible when filed
  • C. Conclusions
  • W805EC, W205ECEDL2, W205GBA2ED and P4075EC appeared equally effective against clinical isolates of MRSA, with MIC90 values of 2 or 4 μg/ml. In addition, based on the ratio of MBC90/MIC90, the four nanoemulsions were bactericidal. As expected, the MRSA were multidrug-resistant, emphasizing the need for new agents for treating MRSA infections. Since nanoemulsions according to the invention also have activity against serious gram-negative pathogens, such as Pseudomonas aeruginosa and Burkholderia spp. (LiPuma et al., “In vitro activities of a novel nanoemulsion against Burkholderia and other multidrug-resistant cystic fibrosis-associated bacterial species,” Antimicrob. Agents Chemother., 53:249-255 (2009)), the nanoemulsions are useful for inhalation treatment/maintenance of cystic fibrosis patients. In addition, the nanoemulsions are useful in treating burn wounds to prevent/treat infections of the pathogenic agents described herein.
  • Example 8
  • The purpose of this example was to determine the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) of an exemplary nanoemulsion (P4075EC) side by side with comparator drugs against clinical isolates of Pseudomonas aeruginosa, Burkholderia cenocepacia, Acinetobater baumanni, Stenotrophomonas maltophilia from patients suffering from cystic fibrosis (CF).
  • Summary of Results for MIC and MBC: P4075EC+EDTA had an MIC range of <4-16, <4-64, 2-8, <1-16 μg CPC/ml, respectively. The addition of EDTA at 5 mM for the Pseudomonas and Burkholderia and 1 mM to Acinetobacter and Stenotrophomonas to each well containing nanoemulsion improved the MIC. MBC data for the nanoemulsion+EDTA were obtained for 20 Pseudomonas, 10 Burkholderia, 10 Acinetobacter and 11 Stenotrophomonas. For three genera of bacteria tested, the data suggested bacteriostatic activity. The MBCs were within 4-fold of the respective MICs for the Stenotrophomonas, consistent with the nanoemulsion having bactericidal activity.
  • Summary of Synergy Results: In addition to the standardized MIC and MBC determinations, checkerboard synergy studies were conducted to evaluate the potential of P4075EC+EDTA to synergize or antagonize traditional antimicrobials commonly used to treat patients with cystic fibrosis. The fractional inhibitory concentration (FIC) index and the fractional bactericidal concentration (FBC) index were determined to judge if a two drug combination was synergistic, antagonistic or indifferent to one another. Ten strains of Burkholderia, 10 strains of Stenotrophomonas and 10 strains of Acinetobacter were tested to determine a shift in MIC when P4075EC+EDTA was in combination with either colistin or tobramycin, two traditional antimicrobials used in the lungs of CF patients to treat chronic lung infections. P4075EC+EDTA in combination with colistin was found to be synergistic for 90% (in terms of the FIC) and 70% (in terms of the FBC) of the Stenotrophomonas strains, but indifferent, only 20% synergy in by the FIC and 0% by the FBC, when in combination with tobramycin. For the Acinetobacter strains, P4075EC+EDTA in combination with colistin was found to be indifferent, only 20% synergy in by the FIC and 0% by the FBC, as well as when in combination with tobramycin, only 10% synergy in by the FIC and 10% by the FBC. For the Burkholderia strains, P4075EC+EDTA in combination with colistin was found to be indifferent, only 30% synergy in by the FIC and 10% by the FBC, but when in combination with tobramycin, 50% synergy in by the FIC and 20% by the FBC.
  • A. Materials and Methods
  • Multidrug-resistant Gram-negative bacteria isolated from CF patients were tested in this example. MICs and MBCs were determined using CLSI guidelines and standard methods M7-A7 and M100-S17. Depending on the genus, MICs were performed in the presence of 1 or 5 mM EDTA, a permeability enhancer of NE activity. Stenotrophomonas and Acintetobacter had 1 mMEDTA in the nanoemulsion. Pseudomonas and Burkholderia had 5 mM EDTA in the nanoemulsion. This was the case during the standard MIC and MBC determination as well as the Checkerboard synergy work and time kill study to be described below. The addition of alamar blue, a redox indicator that yields a colorimetric change in response to metabolic activity, was used to determine the MICs of the NE (P4075EC) because of opacity at higher concentrations.
  • Checkerboard synergy studies were carried out by using the MIC and MBC data collected, microtiter plates with a combination for P4075EC+EDTA and colistin or P4075EC+EDTA and tobramycin.
  • The Fractional Inhibitory Concentration (FIC) Index (see e.g., FIG. 19) examines the ratio of the MIC of a single drug when in combination with another to the MIC of that drug alone. The reduction of the MIC when a drug is in combination results in a fraction that is less than one.

  • X=MIC of Drug in Combination/MIC of Drug along
  • This ratio is calculated for both drug A and drug B. The fractions are added together. The summation is compared to the following ranges:
  • Synergism: Sum of FIC for the two drugs≦0.5;
  • Indifference: Sum of FIC for the two drugs>0.5 to ≦4;
  • Antagonism: Sum of FIC for the two drugs>4.
  • In FIG. 19A, Row D denotes the previously determined MIC used to chose the flanking concentrations of the two drugs being combined. In FIG. 19B, the horizontal slash marks on the X and Y axis indicate typical MIC concentration of each drug alone.
  • The time-kill study was done over a brief time curve of 10, 20, 30 minutes including a 0 minute non-treated control was conducted to fix samples for pictures under the electron microscope.
  • B. Source of isolates
  • 46 isolates were evaluated, all of which were obtained from the University of Maryland-Baltimore School of Dentistry. The isolates received were as follows: 20 Pseudomonas, 11 Burkholderia, 10 Acinetobacter and 5 Stenotrophomonas. The P. aeruginosa isolates had defined lipid A modifications that have been documented in many CF patients. 10 additional isolates of Stenotrophomonas were received from a second source (Eurofins, Virginia). These 10 are the last on the list and are the isolates with the ages of the individuals the samples were taken from. The isolates are summarized in Table 14 below.
  • TABLE 14
    Pseudomonas aeruginosa, Burkholderia cenocepacia, Acinetobacter baumannii,
    Stenotrophomonas maltophilia isolates
    Pseudomonas Aminoarabinose modification Isolate Patient
    aeruginosa NanoBio number Probable colistin resistance Location Identifier
    1 NB1  CF Patient A
    2 NB2  CF Patient A
    3 NB3  + CF Patient B
    4 NB4  + CF Patient B
    5 NB5  + CF Patient C
    6 NB6  + CF Patient C
    7 NB7  + CF Patient D
    8 NB8  + CF Patient D
    9 NB9  CF Patient E
    10  NB10 CF Patient E
    11  NB11 CF Patient F
    12  NB12 CF Patient G
    13  NB13 CF Patient G
    14  NB14 CF Patient H
    15  NB15 CF Patient I
    16  NB16 + CF Patient J
    17  NB17 CF Patient K
    18  NB18 CF Patient L
    19  NB19 CF Patient M
    20  NB20 + CF Patient N
    Burkholderia Isolate Patient
    cenocepacia NanoBio number Genomovar Location Identifier
    1 NB21 B. c. Genomovar I CF Patient A
    2 NB22 B. c. Genomovar I CF Patient B
    3 NB23 B. c. Genomovar III CF Patient C
    4 NB24 B. c. Genomovar III CF Patient D
    5 NB25 B. c. Genomovar III CF Patient E
    6 NB26 B. c. Genomovar III CF Patient F
    7 NB27 B. c. Genomovar III CF Patient G
    8 NB28 B. c. Genomovar IV CF Patient H
    9 NB29 B. c. Genomovar IV CF Patient I
    10  NB30 B. c. Genomovar IV CF Patient J
    11  NB28b B. vietnamienis Genomovar V
    Acinetobacter Isolate Patient
    baumannii NanoBio number Colistin Susceptibility Location Identifier
    1 NB31 Sensitive wound, intra- Unknown
    abdominal
    2 NB32 Sensitive abcess Unknown
    3 NB33 Resistant peritoneal fluid Unknown
    4 NB34 Sensitive sub-clav blood Unknown
    5 NB35 Resistant blood Unknown
    6 NB36 Resistant wound Unknown
    7 NB37 Sensitive tissue Unknown
    8 NB38 Sensitive urine Unknown
    9 NB39 Resistant CSF Unknown
    10  NB40 Resistant urine Unknown
    Isolate Patient
    Stenotrophomonas NanoBio number Location Identifier
    1 NB41 CF Patient A
    2 NB42 CF Patient B
    3 NB43 CF Patient C
    4 NB44 CF Patient D
    5 NB45 CF Patient E
    Date
    Stenotrophomonas Isolation Source
    maltophilia Short Id. Region Year Description Age
    2482274    1 NEW ENGLAND 2006 Sputum 14
    2482270    2 EAST NORTH CENTRAL 2007 Sputum 10
    2482275    3 MOUNTAIN 2007 Sputum 17
    2482269    4 MID ATLANTIC 2008 Sputum 15
    2482273    5 SOUTH ATLANTIC 2008 Sputum 17
    2482276    6 EAST SOUTH CENTRAL 2008 Sputum 9
    2482267    7 WEST NORTH CENTRAL 2008 Sputum 13
    2482271    8 WEST SOUTH CENTRAL 2008 Sputum 4
    2482272    9 PACIFIC 2008 Sputum 16
    2482268    10  SOUTH ATLANTIC 2008 Sputum 1

    C. Source of drugs
  • The drugs and the source thereof used in the drug panel plates are summarized in the table below.
  • TABLE 15
    Drugs used in drug panel plates.
    Ingredient Manufacturer Lot #
    Cefepime USP H0G278
    Colistin USP G-1
    Imipenem USP H0E040
    Levofloxacin Sigma 1333515
    Ceftazidime USP H
    Tobramycin USP L0E077
    Ceftoxin USP J0E038
    Piperacillin USP H
    P407-5EC NanoBio X1138 and X1180
  • The composition of the P407-5EC is provided in Table 16, below. The emulsion was produced by mixing a water immiscible oil phase with an aqueous phase. The base formulation is below and represents the neat emulsion, which was further diluted to the desired %.
  • TABLE 16
    Lot # A0499
    Neat P407 5EC
    Ingredient w/w %
    Sterile distilled water 23.49
    CPC 1.068
    Poloxamer 407 5.92
    Ethanol 6.73
    Soybean oil 62.79
  • D. Preparation of Drug Concentrations
  • The preparation of drug concentration is described below in Tables 17 and 18. Specifically, Table 17 provides details regarding the preparation of the 96-well drug panel plates, and Table 18 provides details regarding the preparation of the 96-well checkerboard synergy plates.
  • TABLE 17
    Details describing the preparation of the 96-well drug panel plates.
    Exp. Stock
    Molecular Concentrations Volume Required
    Weight Potency Starting in Concentration Weight
    Name & Lot # g/mole (ug/mg) at X ug/mL: Solvent/Diluent mL (ug/mL) Needed (mg)
    Cefepime-USP 571.5 865 32 Phos. Buffer 5.00 3200 18.50
    Lot H0G278 pH 6
    Colistin 1163 629 8 water 10.00 800 12.72
    (Polymyxin E)-
    USP Lot G-1
    Imipenem-USP 317.36 929 32 Phos. Buffer 5.00 3200 17.22
    Lot H0E040 pH 7.2
    Levofloxacin- 361.37 999.00 16 ½ volume 10.00 1600 16.02
    Sigma 1333515 water, then 0.1 mol/L
    NaOH
    dropwise to
    dissolve
    Piperacillin- 535.57 984 1024 ½ volume 2.00 102400 208.13
    USP Lot H water, then 0.1 mol/L
    NaOH
    dropwise to
    dissolve
    Tobramycin- 467.52 970 32 water 5.00 3200 16.49
    USP Lot
    L0E077
    Cefoxitin-USP 449.44 992 256 ½ volume 2.00 25600 51.61
    Lot J0E038 water, then 0.1 mol/L
    NaOH
    dropwise to
    dissolve
    Ceftazidime-Lot H 535.57 852 32 Calcium 2.00 3200 7.51
    Carbonate Sln
    10% of the
    weight of
    compound to
    be dissolved
    YELLOW HIGHLIGHT INDICATES POTENCY TAKEN FROM PERCENT PURITY (99% = 999 ug/mg).
    1st Dilute ½, nine times from stock in solvent
    2nd Dilute 1/50 in medium in a respective well of a 12 well trough.
    3rd Transfer 50 uL from each well of the reservoir into the respective well for each agent at each concentration.
    Required
    with or Solvent Stock Concentration Volume Needed Volume
    without and Volume in (mM or of Initial Stock Needed of
    Nanoemulsion EDTA Volume mL ug/mL) (mL) broth (mL)
    X1138 or with 1 mM Water 4.00 512 0.68 3.32
    X1180 = P4075EC
    (@ 6 mg/mL)
    X1138 or with 5 mM Water 4.00 2056 2.74 1.26
    X1180 = P4075EC
    (@ 6 mg/mL)
    ** Because of the high concentration of this test antimicrobial, a 100x stock could not be made.
    The calculations provided a 2x stock that is diluted in broth from the start and through the nine 1/2 dilutions.
    A 1/50 dilution at any time is not required because the dilutions are made directly in broth.
    X mM
    EDTA
    Diluted EDTA
    in Broth Broth Stock
    @ 100x volume Volume Total Volume
    EDTA Stock
    100 800 200 1000
    solution starting 500 0 1000 1000
    at 500 mM
    EDTA used to
    make these
    dilutions.
    For the required concentration of EDTA, a 1/50 dilution of the EDTA 100x stock was made in each well of the 12 well
    trough containing nanoemulsion.
    Only 10 wells of the 12 well trough contain drug, the last two wells are for broth only which set up the negative and
    positive growth controls.
  • TABLE 18
    Details describing the preparation of the 96-well checkerboard synergy plates.
    Required
    with or Solvent Stock Concentration Volume Needed Volume
    without and Volume (mM or of Initial Stock Needed of
    Nanoemulsion EDTA Volume in mL ug/mL) (mL) broth (mL)
    X1180 = P4075EC with 1 or Broth 4.00 Varied on plate
    (@ 6 mg/mL) 5 mM Broth 4.00 for each strain
    of bacteria
    Colistin no Broth 4.00 Varied on plate
    (Polymyxin E)- for each strain
    USP Lot G-1 of bacteria
    Tobramycin- no Broth 4.00 Varied on plate
    USP Lot L0E077 for each strain
    of bacteria
    ** Because of the high concentration of this test antimicrobial, a 100x stock could not be made.
    The calculations provided a 4x stock that is diluted in broth from the start and through the nine ½ dilutions.
  • E. Determination of MICs and MBCs
  • In the 96 well microtiter plate, the inoculated wells were observed after 20 hours of incubation at 35 degrees C. In each row, there is a range from high to low concentrations of the drug respective of the row from left to right, each well ½ the concentration of the adjacent well to the left. The first clear well in the series was called the MIC. This is the concentration of drug that inhibited the growth of the bacteria from turning the broth turbid. Because of the nanoemulsions intrinsic turbidity, alamar blue (CellTiter Blue, Promega) was added for the row containing nanoemulsion at a rate of 20 uL to 100 uL of culture volume. The plate was returned to the incubator for one hour and the first blue well was called the MIC (wells with bacterial growth turned pink at lower concentrations of nanoemulsion). The starting inoculum in each well was generally between 2-7×10̂5 cfu/mL. The specific concentration for starting inoculum was determined for each test plate by sampling the inoculum used to inoculate the microtiter plate at the time it was set up. The exact concentration of bacteria was used to determine the MBC for each drug in the panel for each test plate. The MBC is defined as the concentration of drug that reduced the number of bacteria 99.9%. The MIC is the landmark used to begin sampling 10 uL/well from the MIC plus four wells to the left and put on agar plates, one sample per plate. The agar plates were incubated for 24 hours at 35 degrees C. and colonies were counted. The resulting colony counts were compared to the starting concentration of the inoculum to determine which well, that is which concentration of drug, resulted in a three log drop in cfu/mL.
  • F. Checkerboard Synergy Study
  • The starting inoculum in each well was generally between 2-7×10̂5 cfu/mL. The specific concentration for starting inoculum was determined for each test plate by sampling the inoculum used to inoculate the microtiter plate at the time it was set up. Those exact concentrations of bacteria were used to determine the MBC for each drug in the panel for each test plate. The MBC is defined as the concentration of drug that reduced the number of bacteria 99.9%, a three log drop from the original concentration of bacteria. The MIC is the landmark used to begin sampling 10 uL/well from the MIC plus all wells through to column 11 to the right and put on agar plates, one sample per plate. The agar plates were incubated for 24 hours at 35 degrees C. and colonies were counted. The resulting colony counts were compared to the starting concentration of the inoculum to determine which well, that is which concentration of drug, resulted in a three log drop in cfu/mL.
  • In the 96 well microtiter plate, the inoculated wells were observed after 20 hours of incubation at 35 degrees C. In each row, there is a range from high to low concentrations of the drug respective of the row from right to left, each well ½ the concentration of the adjacent well to the left. Typically, but not always, the rows contained the nanoemulsion, drug A. Going up and down in the columns is drug B and here is the second drug tested in combination going high to low from A to G, respectively. Row H did not contain the second drug for it to act as the control for drug A. Column 1 did not contain any drug A so that column 1 could be the control for drug B. Traditionally, first clear well in the series from right to left in the rows was called the MIC. This is the concentration of drug that inhibited the growth of the bacteria from turning the broth turbid, in this case as indicated by a color change from blue to pink since alamar blue was used to find the metabolic activity to define the MIC. To each well 20 uL of CellTiter Blue from Promega was added. That is 20% of the original culture volume.
  • The well with the MIC and the well with the MBC were noted for each row. All wells noted for the MIC of the two drugs in combination were compared to the MIC of concentration of drug A and drug B each acting alone. For example, a well that contained 2 ug/mL of drug A and 8 ug/mL of drug B would have the concentration of A at 1 ug/mL when in combination divided by 16 ug/mL when acting alone. This renders a ratio of 0.125. Further in this example, 8 ug/mL of drug B would be divided by 32 ug/mL of B acting alone. The ratio for drug B in combination:alone is then 0.25. In this example the FIC index is said to be 0.125 for drug A and 0.25 for drug B, the sum is 0.325 in this example. The definition of synergy is for the sum to be equal to 0.5 or less. Indifference is greater than 0.5 to less than or equal to four. Antagonism is greater than four. See e.g., FIG. 19 for a detailed illustration.
  • G. Time-Kill Study
  • A bacterial suspension of Burkholderia NB 29@O.D. 0.14-0.20 was made up. In a saline tube, 500 uL saline and 500 uL of the bacterial suspension was mixed for the TO, non-treated control, then mixed one to one with TA (Trypton-Azolectin) broth with 5 mM CaCl2 to be diluted and plated as described below. In a treatment tube, added one to one the bacterial suspension to the treatment mix (2×P4075EC at 64 ug/mL CPC and 10 mM EDTA) and after 10, 20 and 30 minutes drew 400 uL at each time point and mixed with 400 uL TA broth with 5 mM CaCl2. Next, 100 uL were drawn to be diluted serially 1/10 in saline to 10−8. 100 uL were then drawn from each dilution tube in the series and plated in triplicate. These plates were incubated at 35° C. overnight and counted to determine the cfu/mL at each time point. Simultaneously at the time the sample of untreated or treated bacteria was added to the neutralization TA solution, a sample of 450 uL was added to 113 uL of glutaraldehyde for each respective time point.
  • H. Scanning Electron Microscopy (SEM)
  • For scanning electron microscopy (SEM), 113 ul of 10% aqueous solution of gluteraldehyde in Sorenson's buffer, pH 7.4, was mixed with 450 uL of the bacterial suspension that underwent the exposure to nanoemulsion. Mixtures were vortexed and placed at 4° C. for at least 18 hours. Table 19 provides the procedure for fixing and staining the samples for scanning electron microscopy.
  • TABLE 19
    Method for fixing and staining samples for SEM
    Step
    1 Samples were fixed in 2.5% glutaraldehyde in Sorenson's buffer, pH 7.4 then
    agitated on a rotary stirrer at each step
    2 Samples were rinsed twice for 15 minutes each in 0.1 Sorensen's buffer
    3 samples were fixed in 1.0% OsO4 in Sorenson's buffer
    4 samples were rinsed twice for 5 minutes each in 0.1 M Sorenson's buffer
    5 Samples were dehydrated for 15 minutes each in each of the following: 30% EtOH,
    50% EtOH, 70% EtOH, 90% EtOH, 100% EtOH, 100% EtOH
    6 Samples were immersed in four, 15 minutes changes of hexamethyldisilazane
    (HMDS)
    7 Samples were removed following the fourth change of HMDS and replaced with just
    enough HMDS to cover tissue. Samples were allowed to evaporate in the hood
    overnight
    8 Samples were mounted on SEM stubs, using the mixture of Colloidal graphite and
    duco cement
    9 Samples were placed in a vacuum desiccator overnight
    10 Samples were sputter-coated with gold using “Polaron” sputter coater
    11 Samples were examined on an “Amray 1910 FE” Sacnning Electron Microscope and
    digitally imaged using “Xstream” imaging software
  • I. Results
  • a. MIC and MBC Data
  • The MIC90/MBC90 values for P4075EC were 8/64 μg/ml for P. aeruginosa, 64/>514 μg/ml for B. cenocepacia, 8/64 μg/ml for A. baumannii and 8/32 μg/ml for S. maltophilia. Colistin had MIC90/MBC90 values of 2/8, >32/>32, 11>16 and >32/>32 for P. aeruginosa, B. cenocepacia, A. baumannii and S. maltophilia, respectively. Cefepime, imipenem, levofloxacin and tobramycin had MIC90/MBC90 values of ≧32/>32, ≧32/>32, 16/16 and >32/>32 μg/ml, respectively, against all strains.
  • b. Synergy Data
  • Ten strains of Burkholderia, Stenotrophomonas and 10 strains of Acinetobacter were tested to determine a shift in MIC when P4075EC+EDTA was in combination with either colistin or tobramycin, two traditional antimicrobials used in the lungs of CF patients to treat chronic lung infections. P4075EC+EDTA in combination with colistin was found to be synergistic for 90% (in terms of the FIC) and 70% (in terms of the FBC) of the Stenotrophomonas strains, but indifferent, only 20% synergy in by the FIC and 0% by the FBC, when in combination with tobramycin. For the Acinetobacter strains, P4075EC+EDTA in combination with colistin was found to be indifferent, only 20% synergy in by the FIC and 0% by the FBC, as well as when in combination with tobramycin, only 10% synergy in by the FIC and 10% by the FBC. For the Burkholderia strains, P4075EC+EDTA in combination with colistin was found to be indifferent, only 30% synergy in by the FIC and 10% by the FBC, but when in combination with tobramycin, 50% synergy in by the FIC and 20% by the FBC.
  • c. Time-Kill Study
  • Time-kill resulted in an overall 4.44 log reduction in cfu/ml from the untreated beginning to the 30 minute time point. Each 10 minute time point had between 1-2 log reduction as follows: from the untreated to the 10 minute point there was a 1.40 log reduction, from the 10 to 20 minute time points there was a 1.91 log reduction and from the to 30 minute time points there was a 1.13 log reduction. See FIGS. 20A-20D.
  • TABLE 20
    A comparison of MIC (μg/ml) of P4075EC and comparator drugs.
    MIC & MBC 50 & 90 CF Pseudomonas Isolates
    All Drug Values in ug/mL
    Cefepime Tobramycin Cefoxitin Colistin Levofloxacin Imipenem
    count Pseudomonas MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC
     1 NB1 2 8 1 16 >256 >256 2 >8 0.5 1 1 8
     2 NB2 0.13 0.125 0.25 2 >256 >256 1 8 0.5 0.5 2 2
     3 NB3 1 2 0.5 4 >256 >256 1 8 0.25 0.5 2 16
     4 NB4 1 1 0.5 4 >256 >256 2 4 0.25 0.5 4 8
     5 NB5 2 4 0.5 4 >256 >256 2 8 0.5 1 2 16
     6 NB6 2 4 0.5 4 >256 >256 2 4 0.5 1 1 2
     7 NB7 2 2 1 >16 >256 >256 2 4 0.5 1 4 16
     8 NB8 2 2 1 8 >256 >256 2 8 0.5 0.5 4 4
     9 NB9 2 2 1 8 >256 >256 2 8 0.5 0.5 4 >32
    10 NB10 2 2 0.5 4 >256 >256 2 2 0.5 0.5 4 8
    11 NB11 16 32 2 4 >256 >256 1 4 8 16 2 8
    12 NB12 4 32 0.5 8 >256 >256 4 8 0.25 1 >32 >32
    13 NB13 4 16 1 4 >256 >256 2 8 0.5 2 8 16
    14 NB14 32 >32 32 >32 >256 >256 2 4 16 >16 >32 >32
    15 NB15 8 >32 0.5 8 >256 >256 2 8 2 4 8 >32
    16 NB16 16 16 2 16 >256 >256 2 4 2 8 8 32
    17 NB17 >32 >32 16 32 >256 >256 1 4 16 16 >32 >32
    18 NB18 >32 >32 16 >32 >256 >256 1 4 4 8 >32 >32
    19 NB19 16 32 2 16 >256 >256 2 4 2 4 >32 >32
    20 NB20 8 8 0.5 8 >256 >256 2 >8 0.5 1 2 16
    MIC 50 Cefepime 2 Tobramycin 1 Cefoxitin >256 Colistin 2 Levofloxacin 0.5 Imipenem 4
    MIC 90 32 16 >256 2 8 >32
    MBC 50 Cefepime 8 Tobramycin 8 Cefoxitin >256 Colistin 4 Levofloxacin 1 Imipenem 16
    MBC 90 >32 >16 >256 8 16 >32
    P407-5EC & 5 mM P407-5EC &
    Ceftazidime Pipercillin EDTA 0 mM EDTA
    count Pseudomonas MIC MBC MIC MBC MIC MBC MIC MBC
     1 NB1 n/a n/a 1024 1024 16.0625 64.25 n/a
     2 NB2 n/a n/a 128 128 8.03125 32.125 n/a
     3 NB3 n/a n/a >1024 >1024 4.015625 16.0625 n/a
     4 NB4 n/a n/a 512 1024 4.015625 16.0625 n/a
     5 NB5 n/a n/a >1024 >1024 8.03125 32.125 n/a
     6 NB6 n/a n/a 1024 >1024 4.015625 16.0625 n/a
     7 NB7 n/a n/a 512 1024 4.015625 16.0625 n/a
     8 NB8 n/a n/a >1024 >1024 4.015625 8.03125 n/a
     9 NB9 n/a n/a 512 >1024 4.015625 64.25 n/a
    10 NB10 n/a n/a 256 512 8.03125 64.25 n/a
    11 NB11 n/a n/a n/a n/a <4.015625 16.0625 n/a
    12 NB12 256 512 n/a n/a <4.015625 16.0625 514 >2056
    13 NB13 512 512 n/a n/a 8.03125 16.0625 514 >2056
    14 NB14 1024 >1024 n/a n/a <4.015625 32.156 4.01563 >64.25
    15 NB15 256 512 n/a n/a <4.015625 16.0625 1028 >2056
    16 NB16 256 >1024 n/a n/a <4.015625 >64.25 2056 >2056
    17 NB17 n/a n/a n/a n/a <4.015625 16.0625 n/a
    18 NB18 n/a n/a n/a n/a <4.015625 <4.015625 n/a
    19 NB19 512 1024 n/a n/a <4.015625 8.03125 1028 >2056
    20 NB20 256 512 n/a n/a 8.03125 8.03125 1028 >2056
    MIC 50 Ceftazidime n/a Pipercillin n/a P407-5EC 4.015625 P407- n/a
    & 5 mM 5EC &
    EDTA 0 mM
    EDTA
    MIC 90 n/a n/a 8.03125 n/a
    MBC 50 Ceftazidime n/a Pipercillin n/a P407-5EC 16.0625 P407- n/a
    & 5 mM 5EC &
    EDTA 0 mM
    EDTA
    MBC 90 n/a n/a 64.25 n/a
    MIC & MBC 50 & 90 CF Burkholderia Isolates
    All Drug Values in ug/mL
    Cefepime Colistin Imipenem Levofloxacin Ceftazidime
    count Burkholderia MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC
    1 NB21 32 >32 >32 >32 16 16 2 2 8 >32
    2 NB22 >32 >32 >32 >32 16 16 1 4 8 >32
    3 NB23 >32 >32 >32 >32 >32 >32 8 >16 >32 >32
    4 NB24 32 >32 >32 >32 32 >32 8 16 4 32
    5 NB25 >32 >32 >32 >32 32 >32 4 8 16 32
    6 NB26 >32 >32 32 >32 >32 >32 8 8 16 32
    7 NB27 >32 >32 >32 >32 16 >32 4 8 8 16
    8 NB29 8 >32 >32 >32 8 16 1 4 4 >32
    9 NB30 8 16 >32 >32 4 4 1 1 2 2
    10  NB28b 4 4 >32 >32 0.5 0.5 1 2 16 16
    MIC 50 Cefepime 32 Colistin >32 Imipenem 16 Levofloxacin 2 Ceftazidime 8
    MIC 90 >32 >32 >32 8 16
    MBC 50 Cefepime >32 Colistin >32 Imipenem 32 Levofloxacin 4 Ceftazidime 32
    MBC 90 >32 >32 >32 >16 >32
    P407-5EC & 5 mM
    Tobramycin Cefoxitin EDTA
    count Burkholderia MIC MBC MIC MBC MIC MBC
    1 NB21 32 >32 256 >256 32.125 514
    2 NB22 >32 >32 128 256 32.125 >514
    3 NB23 >32 >32 >256 >256 <4.015625 64.25
    4 NB24 >32 >32 128 >256 32.125 514
    5 NB25 >32 >32 128 256 64.25 257
    6 NB26 >32 >32 128 256 <4.01563 64.25
    7 NB27 >32 >32 128 >256 32.125 257
    8 NB29 >32 >32 256 >256 16.0625 >514
    9 NB30 32 32 128 128 <4.015625 >64.25
    10  NB28b 1 4 16 16 <4.015625 >64.25
    MIC 50 Tobramycin >32 Cefoxitin 128 P407-5EC 16.0625
    & 5 mM
    EDTA
    MIC 90 >32 256 64.25
    MBC 50 Tobramycin >32 Cefoxitin 256 P407-5EC 128.5
    & 5 mM
    EDTA
    MBC 90 >32 >256 >514
    MIC & MBC 50 & 90 CF Acinetobacter Isolates
    All Drug Values in ug/mL
    Cefepime Colistin Imipenem Levofloxacin Ceftazidime
    count Acinetobacter MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC
    1 NB31 16 16 1 1 1 1 16 16 >32 >32
    2 NB32 32 >32 1 8 32 32 8 8 >32 >32
    3 NB33 >32 >32 1 4 32 >32 8 8 >32 >32
    4 NB34 32 32 1 1 32 32 16 16 32 32
    5 NB35 32 >32 1 4 8 8 >16 >16 >32 >32
    6 NB36 2 2 1 1 0.125 0.125 1 1 2 4
    7 NB37 32 >32 1 >16 4 8 16 16 32 32
    8 NB38 >32 >32 1 >16 32 >32 8 >16 >32 >32
    9 NB39 >32 32 1 8 2 2 16 16 32 >32
    10  NB40 >32 >32 1 >16 2 32 >16 >16 >32 >32
    MIC 50 Cefepime 32 Colistin 1 Imipenem 4 Levofloxacin 16 Ceftazidime >32
    MIC 90 >32 1 32 >16 >32
    MBC 50 Cefepime >32 Colistin 4 Imipenem 8 Levofloxacin 16 Ceftazidime >32
    MBC 90 >32 >16 >32 >16 >32
    P407-5EC &
    Tobramycin Cefoxitin 1 mM EDTA
    count Acinetobacter MIC MBC MIC MBC MIC MBC
    1 NB31 1 1 >256 >256 4 16
    2 NB32 1 4 256 256 4 64
    3 NB33 2 8 >256 >256 4 64
    4 NB34 16 >32 >256 >256 4 8
    5 NB35 2 16 >256 >256 8 32
    6 NB36 0.5 0.5 128 128 2 16
    7 NB37 0.5 2 >256 >256 2 16
    8 NB38 1 16 >256 >256 4 64
    9 NB39 2 4 >256 >256 2 16
    10  NB40 >32 >32 >256 >256 8 >128
    MIC 50 Tobramycin 1 Cefoxitin >256 P407- 4
    5EC & 1 mM
    EDTA
    MIC 90 16 >256 8
    MBC 50 Tobramycin 4 Cefoxitin >256 P407- 16
    5EC & 1 mM
    EDTA
    MBC 90 >32 >256 64
    MIC & MBC 50 & 90 CF Stenotrophomonas Isolates
    All Drug Values in ug/mL
    Cefepime Colistin Imipenem Levofloxacin Ceftazidime
    count Stenotrophomonas MIC MBC MIC MBC MIC MBC MIC MBC MIC MBC
    1 NB41 32 >32 2 32 >32 >32 2 4 >32 >32
    2 NB42 >32 >32 2 8 >32 >32 8 16 8 >32
    3 NB43 >32 >32 16 >32 >32 >32 2 4 >32 >32
    4 NB44 >32 >32 32 >32 >32 >32 8 16 >32 >32
    5 NB45 32 >32 8 16 >32 >32 1 2 >32 >32
    6 Steno 1 >32 >32 >32 >32 >32 >32 1 4 >32 >32
    7 Steno 2 >32 >32 32 >32 32 >32 4 8 >32 >32
    8 Steno 3 >32 >32 >32 >32 >32 >32 1 4 >32 >32
    9 Steno 4 16 >32 1 4 >32 >32 0.5 2 2 16
    10  Steno 6 >32 >32 16 >32 32 32 2 4 32 >32
    11  Steno 8 >32 >32 8 32 >32 >32 2 2 16 >32
    MIC 50 Cefepime >32 Colistin 16 Imipenem >32 Levofloxacin 2 Ceftazidime >32
    MIC 90 >32 >32 >32 8 >32
    MBC 50 Cefepime >32 Colistin >32 Imipenem >32 Levofloxacin 4 Ceftazidime >32
    MBC 90 >32 >32 >32 16 >32
    Acinetobacter
    MIC 50 32 1 4 16 >32
    MIC 90 >32 1 32 >16 >32
    MBC 50 >32 4 8 16 >32
    MBC 90 >32 >16 >32 >16 >32
    Stenotrophomonas
    MIC 50 >32 16 >32 2 >32
    MIC 90 >32 >32 >32 8 >32
    MBC 50 >32 >32 >32 4 >32
    MBC 90 >32 >32 >32 16 >32
    MBC 50 8 4 16 1 Not Tested
    MBC 90 >32 8 >32 16 Not Tested
    Burkholderia
    MIC 50 32 >32 >32 2 8
    MIC 90 >32 >32 >32 8 16
    MBC 50 >32 >32 32 4 32
    MBC 90 >32 >32 >32 >16 >32
    P407-5EC &
    Tobramycin Cefoxitin 1 mM EDTA
    count Stenotrophomonas MIC MBC MIC MBC MIC MBC
    1 NB41 >32 >32 >256 >256 8 8
    2 NB42 32 >32 >256 >256 16 16
    3 NB43 >32 >32 >256 >256 8 16
    4 NB44 >32 >32 256 >256 <1 16
    5 NB45 >32 >32 >256 >256 8 16
    6 Steno 1 >32 >32 >256 >256 8 16
    7 Steno 2 >32 >32 >256 >256 4 16
    8 Steno 3 >32 >32 256 >256 8 32
    9 Steno 4 >32 >32 256 256 2 32
    10  Steno 6 16 32 128 256 8 32
    11  Steno 8 >32 >32 >256 >256 8 16
    MIC 50 Tobramycin >32 Cefoxitin >256 P407- 8
    5EC & 1 mM
    EDTA
    MIC 90 >32 >256 8
    MBC 50 Tobramycin >32 Cefoxitin >256 P407- 16
    5EC & 1 mM
    EDTA
    MBC 90 >32 >256 32
    Acinetobacter
    MIC 50 1 >256 4
    MIC 90 16 >256 8
    MBC 50 4 >256 16
    MBC 90 >32 >256 64
    Stenotrophomonas
    MIC 50 >32 >256 8
    MIC 90 >32 >256 8
    MBC 50 >32 >256 16
    MBC 90 >32 >256 32
    MBC 50 8 >256 16.0625
    MBC 90 >16 >256 64.25
    Burkholderia
    MIC 50 >32 128 16.0625
    MIC 90 >32 256 64.25
    MBC 50 >32 256 128.5
    MBC 90 >32 >256 >514
  • TABLE 21
    Isolate FIC Synergy FBC Synergy Isolate FIC Synergy FBC Synergy
    Checkerboard Synergy Study Results; Acinetobacter
    NB31 Colistin 0.83 indifferent 2.44 indifferent NB31 Tobra 0.98 indifferent 1.50 no
    NB32 Colistin 0.58 indifferent 1.36 indifferent NB32 Tobra 0.88 indifferent 1.79 no
    NB33 Colistin 0.33 yes 2 indifferent NB33 Tobra 1.27 indifferent 1.75 no
    NB34 Colistin 0.46 yes 1.10 indifferent NB34 Tobra 0.41 yes 0.24 yes
    NB35 Colistin 0.72 indifferent 0.97 indifferent NB35 Tobra 1.21 indifferent 1.44 no
    NB36 Colistin 0.56 indifferent 2 indifferent NB36 Tobra 3.30 indifferent 2.29 no
    NB37 Colistin 0.63 indifferent 1.00 indifferent NB37 Tobra 0.75 indifferent 1.72 no
    NB38 Colistin 0.77 indifferent 0.68 indifferent NB38 Tobra 1.05 indifferent 1.57 no
    NB39 Colistin 0.72 indifferent 1.22 indifferent NB39 Tobra 3.30 indifferent 3.52 no
    NB40 Colistin 0.63 indifferent 0.91 indifferent NB40 Tobra 3.01 indifferent 3.01 no
    20%  0% 10% 10%
    Checkerboard Synergy Study Results; Stenotrophomonas
    NB42 Colistin 0.08 yes 1.28 indifferent NB42 Tobra 0.14 yes 2.15 indifferent
    NB43 Colistin 0.08 yes 0.15 yes NB43 Tobra 1.40 indifferent 2.29 indifferent
    NB44 Colistin 1.01 indifferent 0.57 indifferent NB44 Tobra 1.5 indifferent 2.86 indifferent
    NB45 Colistin 0.04 yes 0.45 yes NB45 Tobra 0.97 indifferent 1.50 indifferent
    Steno 1 Colistin 0.08 yes 0.11 yes Steno 1 Tobra 0.79 indifferent 1.57 indifferent
    Steno 2 Colistin 0.09 yes 0.04 yes Steno 2 Tobra 0.04 yes 2.23 indifferent
    Steno 3 Colistin 0.09 yes 0.14 yes Steno 3 Tobra 0.79 indifferent 1.29 indifferent
    Steno 4 Colistin 0.46 yes 1.14 indifferent Steno 4 Tobra 1.72 indifferent 1.61 indifferent
    Steno 6 Colistin 0.09 yes 0.02 yes Steno 6 Tobra 0.59 indifferent 0.89 indifferent
    Steno 8 Colistin 0.09 yes 0.07 yes Steno 8 Tobra 0.63 indifferent 1.15 indifferent
    90% 70% 20%  0%
    Checkerboard Synergy Study Results; Burkholderia
    NB21 Colistin 1.45 indifferent 2.07 indifferent NB21 Tobra 0.27 yes 0.48 yes
    NB22 Colistin 1.37 indifferent 2.29 indifferent NB22 Tobra 0.35 yes 1.23 indifferent
    NB23 Colistin 1.01 indifferent 2.00 indifferent NB23 Tobra 1.06 indifferent 1.09 indifferent
    NB24 Colistin 1.72 indifferent 2.00 indifferent NB24 Tobra 1.31 indifferent 1.36 indifferent
    NB25 Colistin 2.00 indifferent 3.00 indifferent NB25 Tobra 1.45 indifferent 0.99 indifferent
    NB26 Colistin 0.56 yes 0.04 yes NB26 Tobra 0.12 yes 0.92 indifferent
    NB27 Colistin 1.93 indifferent 1.64 indifferent NB27 Tobra 1.44 indifferent 1.27 indifferent
    NB28b Colistin 0.29 yes 1.54 indifferent NB28b Tobra 0.54 yes 1.06 indifferent
    NB29 Colistin 1.61 indifferent 1.68 indifferent NB29 Tobra 0.32 yes 0.07 yes
    NB30 Colistin 0.02 yes 1.86 indifferent NB30 Tobra 0.27 yes 0.54 yes
    30% 10% 50% 30%
  • TABLE 22
    Summary of Synergy Data with Comparison to Original
    MIC/MBC Data of Each Drug Alone
    P4075EC &
    Stenotrophomonas Colistin Tobramycin 1 mM EDTA
    MIC
    50 16 >32 8
    MBC 50 >32 >32 16
    MIC 90 >32 >32 8
    MBC 90 >32 >32 32
    % FIC Synergy 90% 20%
    % FBC Synergy 70%  0%
    Synergy No Interference
    P4075EC &
    Acinetobacter Colistin Tobramycin 1 mM EDTA
    MIC
    50 1 1 4
    MBC 50 4 4 16
    MIC 90 1 16 8
    MBC 90 >16 >32 64
    % FIC Synergy 20% 10%
    % FBC Synergy  0% 10%
    No Interference No Interference
    P4075EC &
    Burkholderia Colistin Tobramycin 5 mM EDTA
    MIC
    50 >32 >32 16
    MBC 50 >32 >32 64
    MIC 90 >32 >32 128
    MBC 90 >32 >32 >514
    % FIC Synergy 30% 50%
    % FBC Synergy 10% 30%
    No Interference No Interference
  • TABLE 23
    Time Kill with additional
    5 mM EDTA
    Burkholderia
    NB29 MIC = 16 Pseudomonas with W205EC at 4 ug/mL + 5 mM EDTA, 1 min resulted in a 2 log drop in count.
    32 ug/mL Burkholderia MIC 50 = 16 ug/mL and 90 = 64 ug/mL
    P4075EC + 5 mM EDTA Starting Bacteria at 0.159 O.D. at 625 nm = ----8.24 × 10{circumflex over ( )}7 cfu/mL
    Set 0 min Set Set Set
    Average (Saline) 8.24E+07 Average 10 min 3.27E+06 Average 20 min 4.00E+04 Average 30 min 2.99E+03
    0 min (Saline) 10 min 20 min 30 min
    Set A cfu/mL Set A cfu/mL Set A cfu/mL Set A cfu/mL
    2.00E+09 0 2.00E+09 0 2.00E+09 0 2.00E+09 0
    2.00E+08 0 2.00E+08 0 2.00E+08 0 2.00E+08 0
    2.00E+07 5 1.00E+08 2.00E+07 0 2.00E+07 0 2.00E+07 0
    2.00E+06 44 8.80E+07 2.00E+06 0 2.00E+06 0 2.00E+06 0
    2.00E+05 128 2.56E+07 2.00E+05 23 4.60E+06 2.00E+05 1 2.00E+05 0
    2.00E+04 TNTC 2.00E+04 180 3.60E+06 2.00E+04 1 2.00E+04 2.00E+04 0
    2.00E+03 Lawn 2.00E+03 TNTC 2.00E+03 19 3.80E+04 2.00E+03 2 4.00E+03
    2.00E+02 Lawn 2.00E+02 TNTC 2.00E+02 151 3.02E+04 2.00E+02 7 1.40E+03
    2.00E+01 Lawn 2.00E+01 Lawn 2.00E+01 TNTC 2.00E+01 134 2.68E+03
    Average 7.12E+07 Average 4.10E+06 Average 2.94E+04 Average 2.69E+03
    Set B cfu/mL Set B cfu/mL Set B cfu/mL Set B cfu/mL
    2.00E+09 0 2.00E+09 0 2.00E+09 0 2.00E+09 0
    2.00E+08 0 2.00E+08 0 2.00E+08 0 2.00E+08 0
    2.00E+07 5 1.00E+08 2.00E+07 0 2.00E+07 0 2.00E+07 0
    2.00E+06 39 7.80E+07 2.00E+06 1 2.00E+06 2.00E+06 0 2.00E+06 0
    2.00E+05 234 4.68E+07 2.00E+05 11 2.20E+06 2.00E+05 1 2.00E+05 0
    2.00E+04 TNTC 2.00E+04 155 3.10E+06 2.00E+04 6 1.20E+05 2.00E+04 0
    2.00E+03 TNTC 2.00E+03 TNTC 2.00E+03 14 2.80E+04 2.00E+03 0
    2.00E+02 Lawn 2.00E+02 Lawn 2.00E+02 177 3.54E+04 2.00E+02 18 3.60E+03
    2.00E+01 Lawn 2.00E+01 Lawn 2.00E+01 TNTC 2.00E+01 128 2.56E+03
    Average 7.49E+07 Average 2.43E+06 Average 6.11E+04 Average 3.08E+03
    Set C cfu/mL Set C cfu/mL Set C cfu/mL Set C cfu/mL
    2.00E+09 0 2.00E+09 0 2.00E+09 0 2.00E+09 0
    2.00E+08 0 2.00E+08 0 2.00E+08 0 2.00E+08 0
    2.00E+07 6 1.20E+08 2.00E+07 1 2.00E+07 0 2.00E+07 0
    2.00E+06 41 8.20E+07 2.00E+06 1 2.00E+06 2.00E+06 0 2.00E+06 0
    2.00E+05 TNTC 2.00E+05 22 4.40E+06 2.00E+05 0 2.00E+05 0
    2.00E+04 TNTC 2.00E+04 172 3.44E+06 2.00E+04 1 2.00E+04 2.00E+04 0
    2.00E+03 Lawn 2.00E+03 TNTC 2.00E+03 18 3.60E+04 2.00E+03 2 4.00E+03
    2.00E+02 Lawn 2.00E+02 TNTC 2.00E+02 164 3.28E+04 2.00E+02 12 2.40E+03
    2.00E+01 Lawn 2.00E+01 Lawn 2.00E+01 TNTC 2.00E+01 161 3.22E+03
    Average 1.01E+08 Average 3.28E+06 Average 2.96E+04 Average 3.20E+03
  • J. Conclusions
  • This examples demonstrates that nanoemulsions, such as the tested P4075EC, are effective against strains that are multidrug-resistant, including colistin-resistant isolates of Burkholderia and Stenotrophomonas. None of the described lipid A modifications in Pseudomonas species impacted the MIC/MBCs with P4075EC. No evidence of antagonism with two major antibiotics, colistin and tobramycin, was observed and in the case of the Stenotrophomonas, synergy was evident. This is valuable because the treatment of patients with CF should not need their normal antibiotic regime suspended in order to use the nanoemulsion, complicating their treatment programs. The SEM images demonstrate the kill on contact mechanism, here in this case a Gram negative bacterium with a reputation of having a tough outer membrane.
  • Example 9 Topical Nanoemulsion Therapy Reduces Bacterial Wound Infection and Inflammation Following Burn Injury Materials and Methods
  • Reagents. Unless otherwise indicated, all reagents were purchased from Sigma-Aldrich Corp. (St. Louis, Mo.).
  • Animals. Male specific pathogen-free Sprague-Dawley rats (Harlan, Indianapolis, Ind.) weighing approximately 250-300 g were used in all experiments. Rats were housed in standard cages and allowed to acclimate to their surroundings for 7 days prior to being used in experiments. They were kept on a 12 hour light cycle and provided with unrestricted access to standard rat chow and water throughout the study. Experiments were performed in accordance with National Institutes of Health guidelines for care and use of animals. Approval for the experimental protocol was obtained from the University of Michigan Animal Care and Use Committee.
  • Burn model. Animals were anesthetized with a 40 mg/kg intraperitoneal (ip) injection of sodium pentobarbital (Nembutal; Abbott Laboratories, North Chicago, Ill.). Dorsal hair was closely clipped and removed using Nair depilatory cream (Church & Dwight Inc., Princeton, N.J.) resulting in a thorough and uniform removal of fur. Each rat was placed in an insulated, custom-made mold, which exposes the dorsal region over 20% of the total body surface area. The burn surface area as a fraction of total body surface area was determined using Meeh's formula: body surface area (cm2)=9.46×(animal weight (g))2/3 (See Gilpin, Burns 22(8):607-611, 1996). Partial thickness scald burn injury was achieved by placing the exposed skin of the rat in a 60° C. water bath for 27 seconds. Sham burn animals received the same treatment except they were immersed in room temperature water (21-24° C.). The burn wound was scrub-débrided with dry sterile gauze and rinsed with 0.9% sterile NaCl. Each animal was resuscitated with 4 mL Ringer's lactate/% total body surface area burn/kg body weight. One half of this fluid volume was given intraperitoneally and half subcutaneously immediately following the burn injury. The burn injured skin was left uncovered to air dry. After drying, an occlusive dressing of sterile TELFA (Kendall Co., Tyco Healthcare Group LP, Mansfield, Mass.) and TEGADERM HP (3M Health Care, St Paul, Minn.) was applied to prevent wound contamination. During experiments each rat was singly housed and received 0.01 mg/kg buprenorphine subcutaneously at the time of burn and at 16 hours for post burn pain control.
  • Local wound treatment. Stock nanoemulsion W205GBA2ED was obtained from NanoBio Corporation (Ann Arbor, Mich.). This nanoemulsion was manufactured by emulsification of super-refined soybean oil and water with surfactants and alcohol as described herein. The resultant droplets had a mean particle diameter of 350 nm. The experimental solution was made by diluting 1 mL of the 60% stock formulation with 4.88 mL sterile saline and adding 120 μL of 1 M ethylenediaminetetraacetic acid (EDTA) giving a final concentration of 10% W205GBA2ED and 20 mM EDTA. A placebo nanoemulsion (W205GBA2ED placebo) compound was manufactured in the same manner as W205GBA2ED, but benzalkonium chloride was omitted from the formulation. 5% Sulfamylon (UDL Laboratories, Inc., Rockford, Ill.) solution was formulated by mixing 50 g of mafenide acetate powder in 1 L of 0.9% sterile saline. The control reagent used was 0.9% sterile saline. Experimental groups consisted of sham, burn, burn+W205GBA2ED, burn+bacteria+saline, burn+bacteria+placebo, burn+bacteria+W205GBA2ED, and burn+bacteria+Sulfamylon. Sixteen hours following burn injury animals were anesthetized with inhaled isoflurane. The occlusive dressing and TELFA was removed. Nanoemulsion (W205GBA2ED), placebo, Sulfamylon or sterile saline was applied in a uniform fashion to the burn wound surface using a spray bottle. Animals in the sham or burn group received no topical treatment, but did undergo dressing change under anesthesia. The burn wound was then redressed with TELFA and a TEGADERM occlusive dressing. This treatment and dressing change was repeated at 24 hours following burn injury.
  • Bacterial culture and inoculation. Pseudomonas aeruginosa isolated from a human burn patient was previously provided by the Department of Pathology at the University of Michigan. This bacterial isolate is sensitive to the topical agent Silvadene and Sulfamylon. A bacterial inoculum was prepared by thawing an aliquot (0.5 mL, stored in 50% skim milk at −80° C.) in 40 mL of Trypticase soy broth (Becton Dickinson, Franklin Lakes, N.J.) and grown overnight at 37° C. with constant shaking at 275 rpm. A sample of the resulting stationary-phase culture was transferred to 35 mL of fresh Trypticase soy broth and incubated for 2.5 hours to reach the log-phase. This subculture was transferred to a 50 mL conical polystyrene tube and centrifuged for 10 minutes at 4° C. and 880 g. The bacterial pellet was washed with 0.9% sterile saline, and resuspended in 10 mL of ice-cold saline. The optical density of the suspension was measured at 620 nm and bacterial concentration (colony forming units (CFU)/mL) calculated using the formula OD620×2.5×108. The bacterial suspension was diluted with 0.9% sterile saline to a final concentration of 1×106 CFU per 100 μL. Eight hours following burn injury animals were anesthetized with inhaled isoflurane. The rats then underwent topical application of 1×106 CFUs of log-phase Pseudomonas aeruginosa in 100 μL of sterile saline pipetted onto a piece of TELFA in a uniform fashion followed by coverage with a TEGADERM occlusive dressing.
  • Tissue harvest. Thirty-two hours after thermal injury the animals were sacrificed and skin tissue samples harvested using sterile technique. Skin samples were used immediately or frozen in liquid nitrogen.
  • Quantitation of bacterial wound infection. A 100 mg piece of excised skin tissue was mechanically homogenized in 1 mL of 0.9 NaCl. This homogenate was then further diluted with 9 mL of sterile saline. Serial dilutions were performed and skin homogenates plated in triplicate on blood agar plates (Becton Dickinson, Franklin Lakes, N.J.). Culture plates were incubated for 24 hours at 37° C. and CFUs counted.
  • Dermal cytokine analysis (ELISA). A 100 mg sample of dorsal skin was homogenized in 1 mL of ice-cold lysis buffer consisting of 50 mL of PBS and protease inhibitor (Complete X, Roche, Indianapolis, Ind.) and 504 of Triton X (Roche). Homogenates were centrifuged at 3000 g for 5 minutes and the supernatants collected and stored frozen at −80° C. until use. Rat IL1-β, IL-6, TNF-α, CINC-1, CINC-3, IL-10 and TGF-β were measured by sandwich enzyme-linked immunosorbent assay (ELISA) using antibodies and reagents from R&D Systems, Inc. (Minneapolis, Minn.). The assay was carried out in 96-well microplates (Immunoplate Maxisorb, Nunc, Neptune, N.J.) according to the kit instructions and samples were read using a microplate reader (Biotek Instruments, Winooski, Vt.) at 450 nm with a wavelength correction of 540 nm. Cytokine concentrations were determined using the plate reader software and a 7-point standard curve. Results were adjusted for previous dilution and expressed as pg/mL.
  • Detection of neutrophil sequestration (Myeloperoxidase assay). 100 mg of skin tissue was mechanically homogenized in 1 mL ice cold potassium phosphate buffer consisting of 115 mM monobasic potassium phosphate (Sigma Aldrich, Milwaukee, Wis.). Homogenates were centrifuged at 3000 g for 10 min at 4° C., the supernatants were removed and the pellets were re-suspended in 1 mL C-TAB buffer consisting of dibasic potassium phosphate, cetyltrimethylammonium bromide, and acetic acid (Sigma Aldrich, Milwaukee, Wis.). The suspensions were sonicated (Branson Sonifier 250, Danbury, Conn.) on ice for 40 seconds. Homogenates were centrifuged at 3000 g for 10 min at 4° C. and the supernatant collected. Supernatants were incubated in 60° C. water bath for 2 hours (Shaker Bath, 2568; Form a Scientific, Marietta, Ohio). Samples were stored at −80° C. until needed or assayed immediately.
  • 20 μL standards (Calbiochem, Gibbstown, N.J.) or samples were added to a 96-well immunosorbent micro-plates (NUNC, Rochester, N.Y.), followed by the addition of 155 μL of 20 mM TMB/DMF consisting of 3,3′,5,5′-tetramethylbenzidine/N,N-dimethylformamide in 115 mM potassium phosphate buffer (Fischer Scientific, Pittsburgh, Pa.) to each well. The samples were mixed well, after which 20 μL of 3 mM H2O2 was rapidly added to each well. The reaction was stopped immediately by adding 504/well of 0.061 mg/mL Catalase (Roche, Indianapolis, Ind.). The plates were read using a microplate reader at 620 nm. Myeloperoxidase (MPO) concentrations were calculated using a linear standard curve and adjusted for previous dilution. The final concentrations were expressed as μg/mL.
  • Determination of dermal capillary leak and tissue edema (Evans blue). Animals were anesthetized 90 minutes before tissue harvest. 50 mg/kg body weight of 10% Evans blue (Merck KgaA, Darmstadt, Germany) was injected ip into the burned animal at time 30.5 hours following thermal injury. At the tissue harvest time point animals were exsanguinated by incision of the inferior vena cava. Systemic Evans blue was washed out by inserting a 20 G angiocatheter into the apex of the left ventricle past the aortic valve and into the ascending aorta. A total of four times the blood volume (7.46 mL/100 g body weight) of 0.9 NaCl with 100 units/mL heparin was used to flush the vasculature and administered via a perfusion pump at a constant flow rate. By the end of the perfusing period the effluent from the right atrium had turned clear. Dorsal skin samples were harvested and a 100 mg sample was placed in 4 mL 99.5% formamide in polyethylene tubes. Tubes were placed on a shaker at room temperature for 48 hours for Evans blue extraction. Supernatants were collected and the absorbance read on a microplate reader at 620 nm. Concentrations were calculated from an Evans blue in formamide standard curve. Results are expressed as micrograms of Evans blue per mg of skin tissue.
  • Histology. Skin samples were fixed in 10% buffered formalin and embedded in paraffin. Eight μm thick sections were affixed to slides, deparaffinized, and stained with hematoxylin and eosin to assess morphologic changes.
  • Detection of hair follicle cell apoptosis (TUNEL assay). Animals were anesthetized and underwent creation of a 20% partial thickness scald burn wound or sham injury.
  • Treatment groups consisted of sham, burn+saline, burn+placebo, and burn+W205GBA2ED. Treatment and dressing changes were performed at 0 and 8 hours post-burn. No bacterial infection was created in this experiment. Full-thickness skin samples were taken from three locations across the entire burn wound at 12, and 24 hours post thermal injury for determination of hair follicle cell apoptosis. There were four animals per treatment group per time sample.
  • Apoptosis was detected in situ with fluorescein based labeling of DNA strand breaks using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay (ApopTag, CHEMICON International, Inc, Temecula, Calif.). The three fresh skin samples for each animal were placed in disposable vinyl cryomolds filled with optimal cutting temperature compound (Sakura Finetek, U.S.A., Inc., Torrance, Calif.), and frozen at −80° C. until ready for use. Frozen embedded skin specimens were cut into 4-mm-thick serial sections in a cryostat and collected on Superfrost Plus glass slides (Fisher Scientific, Pittsburgh, Pa.). Sections were fixed in 1% paraformaldehyde in PBS overnight at 4° C. for indirect immunofluorescence according to the manufacturer's instructions. After 2 washes with PBS, sections were post-fixed in a 2:1 solution of ethanol:acetic acid for 5 minutes at −20° C. Following 2 additional washes with PBS, sections were incubated in equilibrium buffer for 5 minutes. Sections were then incubated with terminal deoxynucleotidyl transferase enzyme in a humidified chamber for 1 h at 37° C. The reaction was terminated by rinsing the sections in a stop/wash buffer. Sections were incubated in a humidified chamber at room temperature with antidigoxigenin fluorescein (fluorescein isothiocyanate, FITC) for 30 min and rinsed 3 times in PBS. After aspiration, sections were washed once with water and coverslips applied using ProLong Gold Antifade (Molecular Probes, Inc, Eugene, Oreg.), which included 4′ 6-diamidino-2-phenylindole dihydrochloride (DAPI) counterstaining.
  • The TUNEL assay slides were blinded to groups, and under the microscope appropriate hair follicle cells in a randomly chosen high-power field were identified. Appropriate hair follicles for analysis were those sectioned in the mid-sagittal or mid-coronal plane. A total of 3-6 hair follicles were selected from among the three skin samples present on a slide. Fluorescent-labeled TUNEL slides were captured digitally at identical time post-labeling to control fading of fluorescence using an Olympus BX-51 fluorescence microscope at fixed image capture settings and 40× magnification. Each hair follicle was selected and first digitally captured by visualizing counterstained nuclei present using the DAPI excitation/emission channel. Then for each hair follicle analyzed, the excitation/filter channel was changed to visualize the fluorescein-labeled TUNEL-positive cells, and images again digitally captured. Within the captured images a region of interest (ROI) was digitally defined, set to include only hair follicle cells and exclude bright fluorescing hair shafts and surrounding cells (NIH Image J software, NIH, Bethesda, Md.). Fluorescence of TUNEL-positive cells was quantified, normalized to ROI size, and expressed as pixels/area fraction, controlling for differences in ROI size.
  • Statistical Methods. Statistical analysis and graphs were performed using GraphPad Prism 5.0 software (GraphPad Software, La Jolla, Calif.). Results are presented as mean values±the SEM unless otherwise noted. Continuous variables were analyzed using an unpaired two-tailed Student's t-test and/or One-way ANOVA followed by Tukeys post-test comparisons. The Kruskal-Wallis test with Dunn's multiple comparisons was used to evaluate differences in medians for data with a non-parametric distribution. Discrete variables were compared using Fisher's exact test. Statistical significance was defined as a p-value<0.05.
  • Topical Application of Nanoemulsion Reduces P. aeruginosa Growth in Burn Wounds
  • Animals treated with nanoemulsion had a decreased mean (6.5×104 vs. 7.9×107, p=0.07) and median (0 vs. 4.4×106, p<0.05) number of CFUs of bacteria per gram of skin tissue when compared to the saline treated controls (See FIG. 21) A similar reduction in skin bacterial counts was found for W205GBA2ED treated animals vs. those treated with the W205GBA2ED placebo (mean: 6.5×104 vs. 5.5×106, p=0.02). When performing quantitative wound culture on clinical tissue samples a positive result is generally considered to be growth of organisms at greater than 1×105 CFUs per g of tissue (See, e.g., Neal et al., J Burn Care Rehabil 2:35-39, 1981; Taddonio et al., Burns Incl Therm Inj 14(3):180-184, 1988; Uppal et al., Burns 33(4):460-463, 2007). Using these criteria, 29 of 32 animals in the control group exhibited evidence of a positive quantitative wound culture and only 3 of 23 animals in the nanoemulsion group demonstrated proof of this level of wound infection (91% vs. 13%, p<0.0001, See Table 24, below). The Sulfamylon treated animals also demonstrated a significant reduction in the median wound bacterial level when compared to the saline controls (3×104 vs. 4.4×106, p<0.05). Treatment with nanoemulsion or Sulfamylon produced a similar reduction in the level of Pseudomonas cultured from the burn wound when compared to the saline treated animals. However, there was no statistically significant difference between the placebo and Sulfamylon groups whereas there was a difference for the W205GBA2ED group compared to the W205GBA2ED placebo.
  • TABLE 24
    Quantitative Wound Culture Results
    Saline Placebo NB-201 Sulfamylon
    Total Animals, n 32 12 23 10
    Animals with 29 (91) 9 (75) 3 (13)* 2 (20)*
    CFU's/g > 1 × 105,
    n (%)
    *p < 0.05, vs. saline control, Fishers exact test
  • Nanoemulsion Treatment Following Burn Injury Attenuates Dermal Pro-Inflammatory Cytokine Levels
  • Scald injury resulting in a partial thickness burn produced differences in dermal levels of IL-1β and cytokine-induced neutrophil chemoattractant-3 (CINC-3) within skin homogenates obtained 32 hours post-injury compared to sham injured animals (See FIGS. 22A and E). Treatment with W205GBA2ED at 16 and 24 hours post-burn reduced the dermal level of these two inflammatory mediators back down to the baseline (sham) in the absence of bacterial infection. In experiments where a bacterial wound infection was not created, a difference in neutrophil sequestration as measured by myeloperoxidase assay was not observed despite the rise in the rat CXC chemokine CINC-3 within burned skin. A difference was found between all three groups (sham, burn, and burn+W205GBA2ED) for CINC-1 (p=0.04, ANOVA), however the values for intergroup comparison did not reach statistical significance.
  • Nanoemulsion Treatment Following Burn Wound Infection with P. aeruginosa Attenuates Dermal Cytokine Levels and Results in Reduced Neutrophil Sequestration
  • Skin homogenates from the nanoemulsion treated group had levels of IL-1β and IL-6 that were considerably diminished when compared to the levels measured in the saline treated animals (See FIGS. 23A and B). There was no statistically significant difference seen in the level of TNF-α between the two experimental groups of animals (See FIGS. 22 C and D). Treatment of the infected burn wound with Sulfamylon did not result in any significant alteration of dermal levels of the measured proinflammatory cytokines (IL-1β, IL-6, TNF-α, CINC-1 or CINC-3) when compared to controls. Treatment with either W205GBA2ED or Sulfamylon reduced the level of myeloperoxidase found in the infected burn wound at 32 hours post-injury. Thus, in some embodiments, treatment with a nanoemulsion and/or antimicrobial reduces the level of neutrophil sequestration into a partial thickness burn wound.
  • Burn injury caused a rise in the level of the anti-inflammatory cytokine TGF-β, but not IL-10 when compared to the sham injured animals (See FIG. 24). W205GBA2ED treatment reduced the amount of TGF-β present in the infected burn wound as compared to the level found in the burn wound alone. Accordingly, in some embodiments, the present invention provides that W205GBA2ED can be utilized to reduce acute burn wound dermal inflammation, and in further embodiments, can be utilized to reduce the eventual immunosuppression created by thermal injury.
  • On histological examination of skin from the saline control animals, there is loss of most of the epidermis and a diffuse cellular infiltrate in the subepidermal region, extending into the lower dermal connective tissue in which collagen fibrils are separated by the infiltrating leukocytes and edema fluid (See FIG. 25A). At a higher power, the cellular infiltrate between the collagen bundles consists almost entirely of neutrophils. Edema fluid causes separation of the collagen fibrils. In FIG. 25B, the skin was subjected to thermal injury followed by application of P. aeruginosa after which the nanoemulsion was topically applied to the burned area. The keratin layers of the epidermis are separating and some of the keratin has been lost. There is a barely detectable intradermal presence of neutrophils together with neutrophils that are adhering to the wall of a venule, which has been longitudinally sectioned (in the center of the microphotograph). The changes in this microphotograph are substantially less extensive than those seen in FIG. 25A.
  • Quantification of Capillary Leak and Tissue Edema
  • Burn wounds are associated with significant levels of capillary leak. This can lead to depletion of the intravascular volume and a need for large amounts of intravenous crystalloid fluid administration. To assess whether therapeutic treatment with a nanoemulsion reduced capillary leak in conjunction with reducing inflammation, an Evans blue assay was utilized to measure vascular permeability. Quantitative measurement of the amount of this dye leaching out of the blood stream and into the skin tissue revealed that the nanoemulsion treated animals had less evidence of post-burn capillary leak and tissue edema than the saline treated controls (See FIG. 26).
  • Treatment with Nanoemulsion Reduces Burn Induced Hair-Follicle Apoptosis
  • Dermal apoptosis occurs in the hair follicle cells following thermal injury. Using a fluorescence labeled TUNEL assay the burn wounds treated with saline showed evidence of intense FITC-TUNEL positive cells which appear green (See FIG. 27). The DAPI nuclear stain allows identification of coronal or sagittally sectioned hair follicles with the cells staining blue. FITC-TUNEL positive cells appear green and are representative of apoptotic cells. In the merged images, the apoptotic hair follicle cells are evident in slides from the burn+saline animals and these changes are diminished in the burn+W205GBA2ED treated animals. Counting the pixels of TUNEL positive cells within a hair follicle region of interest allowed quantification of the reduction in hair follicle cell apoptosis by treatment with topical nanoemulsion (See FIG. 28). The saline treated control animals had an increased amount of TUNEL positive cells when compared to the sham burn animals. Both the W205GBA2ED and placebo treatment resulted in a decrease in hair follicle cell apoptosis following partial thickness burn injury in tissue harvested 12 hours following thermal injury. This difference was not evident in the dermal skin sampled at 24 hours post-burn. Thus, in some embodiments, the present invention provides a method of reducing apoptotic cell death in hair follicles in the early post-burn period comprising administering a nanoemulsion (e.g., W205GBA2ED) to a burn wound during an early post-burn period.
  • Example 10 Immunogenic Compositions Comprising Nanoemulsion and Burkholderia Immunogen and Methods of Inducing Protective Immunity Against Burkholderia Species in a Subject Materials and Methods
  • B. cenocepacia Strain K56-2 Stock Maintenance and Culture. Burkholderia cenocepacia strain K56-2 was generously provided by Dr. Pam Sokol (University of Calgary). K56-2 is a clinical isolate and has been used for Burkholderia molecular microbiology and genomic studies (See, e.g., Goldberg, 2007 Burkholderia Molecular Microbiology and Genomics. Gent, Belgium: Taylor & Francis). It is a representative of the transmissible and virulent B. cenocepacia ET12 lineage (See, e.g., Johnson et al., 1994, J Clin Microbiol 32, 924-930; Saijan et al., 2008, Infect Immun 76, 5447-5455). Burkholderia multivorans ATCC 17616 was used for cross-strain neutralization assays. Both K56-2 and ATCC 17616 strains were stored at −80° C. in Luria-Bertani broth with 15% glycerol and recovered from frozen stock on brain-heart infusion agar overnight at 37° C.
  • B. cenocepacia Outer Membrane Protein (OMP) Preparation. An overnight culture of K56-2 in brain-heart infusion media was centrifuged at 3500 rpm for 20 minutes. The cell pellet was washed several times with PBS (pH 7.4), re-suspended in 1 mM EDTA in PBS (pH 8.0), then incubated for 30 minutes at room temperature. Post-incubation, the bacteria were passed several times through a 26-gauge needle (Becton Dickinson) using high pressure. Cell lysis was achieved with Triton X-100 (Sigma) added to a final concentration of 2% and incubated for 10 minutes at room temperature. Mechanical separation was performed with multiple rounds of sonication (1 minute each). The sonicated lysates were then centrifuged twice for 10 minutes at 6000×g (Beckman Optima XL-100 k ultracentrifuge, 25° C.) with the supernatant retained after each spin. Following the second centrifugation, the supernatant was centrifuged at 100,000×g for 1 hour at 4° C. The resulting pellet was resuspended in endotoxin-free PBS. The rough OMP preparation was purified (endotoxin-depleted OMP) with an endotoxin-removal column (Pierce) according to the manufacturer's instructions. The flow-through fractions were stored at −80° C. until used.
  • OMP Analysis. The protein contained within the OMP preparation was quantified using BCA Assay (Pierce). Western blotting and silver staining were performed according to established protocol as described previously (See, e.g., Makidon et al., 2008, PLoS ONE 3, e2954). To quantify endotoxin contaminate, OMP was analyzed using the LAL Kinetic-QCL (Lonza). Endotoxin was detected at 1.93 endotoxin units/mg in the endotoxin-depleted OMP preparation. DNA contaminant removal was verified by agarose-EtBr gel electrophoresis and imaged on a UV table (See FIG. 29A). Oligonucleotides contaminants were not detected in either the crude OMP or the endotoxin-depleted OMP preparations.
  • OMP Sequencing and Verification.
  • Sample Preparation. The protein sample was separated by SDS-PAGE (See FIG. 29B). The protein band immunostained with the highest intensity (˜17 KDa) on western blot (See FIG. 29C) was excised and subjected to in-gel digestion. In-gel digestion was performed at the Protein Structure Facility at the University of Michigan according to procedures by Rosenfeld et al. and Hellman et al. See, e.g., Hellman et al., 1995, Analytical Biochemistry 224, 451-455; Rosenfeld et al., 1992, Analytical Biochemistry 203, 173-179). A gel slice containing 5-ρmol bovine serum albumin (BSA) was analyzed in parallel as a positive control.
  • Mass Spectrometry. HPLC-Electrospray Ionization (ESI) Tandem Mass Spectrometry (MS/MS) was performed on a Q-tof premier mass spectrometer (Waters Inc) fitted with a nanospray source (Waters Inc.). The mass spectrometer was calibrated with a mass accuracy within 3 ppm. On-line capillary HPLC was performed using a Waters UPLC system with an Atlantis C18 column (Waters, 100-mm inner diameter, 100-mm length, 3-mm particle size). Digests were desalted using an online trapping column (Waters, Symmetry C18, 180-mm inner diameter, 20-mm length, 5-mm particle size) before being loaded onto the Atlantis column. A data-dependent tandem mass spectrometry approach was utilized to identify peptides in which a full scan spectrum (survey scan) was used, followed by collision-induced dissociation (CID) mass spectra of all ions in the survey scan with a peak intensity rising above 20 counts per second. The survey scan was acquired in V+mode over a mass range of 50-2000 Da with lockmass correction (Gu-Fibrinopeptide B, (M+H)2+: 785.8426) and charge state peak selection (2+, 3+ and 4+). The MS/MS scans were acquired for 5 seconds with collision energy control by charge state recognition.
  • Data Analysis and Bioinformatics. Tandem mass spectra were acquired using Mass Lynx software (Version 4.1). Raw data files were first processed for lockmass correction, noise reduction, centering, and deisotoping using the Protein Lynx Global Server software Ver. 4.25 (Waters Inc.). The generated peaklist files containing the fragment mass spectra were subjected to database searches using the ProteinLynxGlobal Server and Mascot (Matrix Science Inc., Boston, Mass.) search engines and Swiss Prot and NCBI databases, as well as the Burkholderia cenocepacia database downloaded from the Sanger Institute web site.
  • Preparation of Nanoemulsion-based OMP (OMP-NE) vaccine. Nanoemulsion (W805EC: 5 vol. % of TWEEN 80, 8 vol. % of ethanol, 1 vol. % of CPC, 64 vol. % of soybean oil) and 22 vol. % of DiH2O) was provided by NanoBio Corporation (Ann Arbor, Mich.), the droplets having a particle diameter of 200-400 nm. OMP-NE formulations were prepared by vigorously mixing the endotoxin-depleted OMP preparation (See FIG. 29B, Lane F) with concentrated NE, using PBS as the diluent. For the intranasal immunizations, OMP-NEs were formulated to contain either 0.25 μg/mL or 0.75 μg/μL OMPs mixed in 20% (v/v) NE.
  • Animals and immunization procedures. Pathogen-free, outbred CD-1 mice (females 6-8 weeks old) were purchased from Charles River Laboratories. The mice were housed in accordance to the standards of the American Association for Accreditation of Laboratory Animal Care. The use of these mice was approved by the University of Michigan University Committee on Use and Care of Animals (UCUCA). The mice (n=10 per group) were vaccinated with two administrations of either OMP-NE vaccines four weeks apart. Intranasal (i.n.) immunizations were performed in mice anaesthetized with isoflurane using the IMPAC6 anesthesia delivery system. The anesthetized animals were held in a supine position and 20 μL (10 μL/nare) of OMP-NE vaccine was administered slowly to the nares using a micropipette tip. Mice were immunized with either 15 μg OMP with NE, 15 μg OMP in PBS, 5 μg OMP with NE, or 5 μg OMP in PBS.
  • Phlebotomy, bronchoalveolar lavage, and splenocyte collection. Blood was collected from the saphenous vein every 21 days throughout the duration of the study. The terminal sample was obtained by cardiac puncture immediately following euthanasia. Whole blood samples were separated by centrifugation at 3500 rpm (15 minutes) following coagulation. The serum samples were stored at −20° C. until analyzed. Bronchoalveolar lavage (BAL) fluid was obtained from the mice immediately following euthanasia as described (See Makidon et al., 2008, PLoS ONE 3, e2954). In vitro measurement of the cytokine response was determined using the spleens of the vaccinated mice. Spleens were mechanically disrupted to obtain single-cell splenocyte suspension in PBS. Red blood cells were lysed with ACK buffer (150 mM NH4Cl, 10 mM KHCO3, 0.1 mM Na2EDTA) and removed by washing the cell suspension twice in PBS. The splenocytes were then resuspended in RPMI 1640 medium supplemented with 2% FBS, 200 nM L-glutamine, and penicillin/streptomycin (100 U/mL and 100 mg/mL).
  • Enzyme-linked immunosorbent assay (ELISA). Serum IgG and mucosal secretory IgA (sIgA) were determined by ELISA against endotoxin-depleted OMP. The OMP was prepared at 15 μg/mL in coating buffer (Sigma) and 100 μL was applied per well to Polysorb plates (Nunc) for a 4° C. overnight incubation. Serum was serially diluted in 0.1% BSA/PBS. Either diluted serum or undiluted BAL was then added to the plates for an overnight incubation at 4° C. Following a 16 hour incubation, the plates were washed and an alkaline phosphatase conjugated anti-mouse IgG (H&L), IgG1, IgG2a, IgG2b, sIgA (a chain-specific) (Rockland Immunochemicals, Inc) antibody was added at 1:2000 or 1:1000 in 0.1% BSA/PBS. The plates were read at OD405 and end titers based on naïve animal levels. Levels of sIgA were normalized to total protein content.
  • Analysis of antigens recognized by serum IgG. In addition to western blot analysis, serum lipoprotein-specific and lipopolysaccharide-specific IgG antibodies in serum were measured using ELISA. Briefly, 96-well Polysorb assay plates (Nunc) were coated with 0.05 μg/well of crude OMP preparation, endotoxin-depleted OMP, or LPS (List Biological Laboratories). Serum was collected from mice immunized with OMP+NE 8 weeks following prime vaccination. The serum was serially diluted in 0.1% BSA/PBS and then added to the coated wells. The ELISA procedure was performed as described supra. To evaluate the immunogenic contribution of protein and LPS contained within the OMP-based vaccine, an enzymatic protein digest was performed using 250 μg of PCR-grade recombinant proteinase K (Roche) in 100 μl of either the crude OMP preparation or the endotoxin-depleted OMP preparation. Proteinase K was incubated with the OMP preparations for 16 hours at room temperature. The protein digestion was verified using a silver-stained SDS-PAGE. LPS and OMP-protein specific epitopes were evaluated using western blot analysis probing with serum from mice vaccinated with either OMP-NE or OMP in PBS, or probing with an anti-Pseudomonas mallei LPS antibody (GeneTex).
  • Analysis of cytokine expression. Freshly isolated mouse murine splenocytes were seeded at 4×106 cells/mL (RPMI 1640, 2% FBS) onto a 24-well plate and incubated for 72 hours with OMPs (15 μg/mL) or PHA-P mitogen (2 μg/mL) as a control. Cell culture supernatants were collected and analyzed for the presence of cytokines using LUMINEX multi-analyte profiling beads (LUMINEX Corporation, Austin, Tex.), according to the manufacturer's instructions.
  • Measurement of serum antibody neutralization. B. cenocepacia strain K56-2 and B. multivorans strain ATCC 17616 antibody-complement neutralization activity was assayed using serum harvested from mice 6 weeks following primary i.n. vaccination. Twenty-five μL of serum (serially diluted 1×, 2×, 3×, 4×, or 5×) was added to 25 μL of a solution containing 1×103 K56-2 or B. multivorans supplementing MH medium in microculture plates (Nunc). The plates were incubated at 37° C. for 48 hours. Controls included 25 μL serum with 25 μL of sterile 1×PBS and 50 μL of MH medium containing 1×103 bacteria. Following incubation, the wells were mixed and 10 μL of solution was plated on Burkholderia selective agar (BCSA) media (See Henry et al., 1997, J Clin Microbiol 35, 614-619) on polystyrene culture plates (Fisherbrand) then incubated 72 hours at 37° C. prior to manual colony forming units (cfu) enumeration.
  • Pulmonary bacterial challenge. Bacterial challenge studies were performed in immunized mice (n=5 per group) or non-vaccinated control mice (n=10) 10 weeks following primary vaccination using a modified version of the chronic pulmonary model of B. cepacia infection described (See Bertot et al., 2007, Infect Immun 75, 2740-2752; Chu et al., 2002 Infect Immun 70, 2715-2720; Chu et al., 2004, Infect Immun 72, 6142-6147). Briefly, 5×107 of K56-2 suspended in 85 μL of PBS was instilled through a trans-tracheal catheter extending to the bifurcation of the main-stem bronchi in anesthetized mice. The mice were maintained for a period of 6 days. Prior to the challenge study, the mice were non-surgically implanted with programmable temperature transponders (IPTT-3000, Bio Medic Data Systems, Inc.) for non-invasive subcutaneous temperature measurement with a handheld portable scanner (DAS-6002, Bio Medic Data Systems, Inc.). In-life analysis included monitoring the clinical status of each animal by evaluating the following metrics: body weight, body temperature, food consumption, and activity. Immediately following euthanasia, pulmonary tissues and the spleen were collected in sterile conditions and each organ was placed into individual containers containing 1 mL of 1×PBS (Mediatec Inc). The tissue was then homogenized using a Tissue TEAROR (Biospec Products Inc.) for 50 seconds on ice. Serial dilutions (1:102 to 1:108) of the homogenate were plated onto separated BCSA plates. All plates were then incubated for 72 hours at 37° C. prior to manual cfu enumeration. Whole blood was collected in tubes containing EDTA (BD) immediately following euthanasia. Anti-coagulated blood was processed to determine total peripheral lymphocytes and mononuclear lymphocytes by the Animal Diagnostic Laboratory at the University of Michigan, using a HEMAVETH 950 hematology analyzer (Drew Scientific, Inc.) in accordance to manufacturer's recommendation.
  • Statistics. Antibody end-titer results are expressed as mean±standard error of the mean (SEM) or ±standard deviation (SD). The statistical significance was determined by ANOVA (analysis of variance) using the Student t and Fisher exact two-tailed tests. For the bacterial challenge studies, the distribution of the colonization was highly skewed, and therefore the data was transformed (natural log transformation) for data normalization. The normalized mean bacterial response in the lung and spleen were compared using an independent samples test with a two sided p-value. p<0.05 was considered to be statistically significant. All analyses were done with 95% confidence limits.
  • Identification of Immunoreactive Proteins
  • Immunoreactive OMP proteins were identified via silver stain (See FIG. 29B) and western blot (See FIGS. 29C and D). Major reactive bands were documented at 62, 45, 17, and 10 KDa in both cases; however, the intensity of the bands was much higher with serum from mice immunized with the NE-based vaccine (See FIG. 29C) as compared to the blot probed with an equivalent serum dilution from mice immunized with the OMP in PBS preparation (See FIG. 29D).
  • The band with the highest intensity was located at ˜17 KDa (See FIG. 29C). For the purpose of identification, the 17 KDa band was isolated from the gel and identified by MALDI-TOF analysis. The protein was identified as Burkholderia cepacia outer membrane lipoprotein A (OmpA) with a MW of 16.396 KDa (See, e.g., Ortega et al., 2005 J Bacteriol 187, 1324-1333; Plesa et al., 2004, J Med Microbiol 53, 389-398). Sequence analysis using National Center for Biotechnology Information (NCBI) protein BLAST confirmed that these proteins are present in other closely related Burkholderia species (See Table 25, below). A high degree of protein homology (87.9% sequence homology) was identified in numerous strains of Burkholderia and Ralstonia organisms and highly conserved amino acids from the OmpA family residues are present in the 17 KDa protein sequence (See Table 25). Thus, the present invention identified and provides immunoreactive OMP proteins from B. cepacia as well as proteins from related strains of Burkholderia species displaying a high degree of homology to the identified immunoreactive OMP proteins from B. cepacia (e.g., that find use in immunogenic compositions comprising a nanoemulsion (e.g., that are administered to a subject in order generate a specific immune response in the subject toward the immunogenic composition comprising nanoemulsion and immunoreactive OMP proteins or homologues)).
  • TABLE 25
    Protein sequence alignment of OmpA-like protein family.
    IDENTITY
    ORGANISMS Omp-A LIKE SEQUENCES ALIGNMENT (%)
    B. CENOCEPACIA LDDKANAGAVSTQPSADNVAQVNVDPLNDPNSPLAKRSIYFDFDSYSVKDEYQPLLQQHAQYLKSHP 100
    QRHVLIQGNTDERGTSEYNLALGQKRAEAVRRALALLGVADSQMEAVS
    LGKEKPLATGHDEASWAQNRRADLVYQQ
    B. DOLOSA LDDKANAGAVSTQPSADNVAQVNVDPLNDPNSPLAKRSIYFDFDSYSVKDEYQPLLQQHAQYL 100
    KSHPQRHVLIQGNTDERGTSEYNLALGQKRAEAVRRALALLGVADSQMEAVSLGKEKP
    LATGHDEASWAQNRRADLVYQQ
    B. MULTIVORANS LDDKANAGA+STQPSADNVAQVNVDPLNDPNSPLAKRSIYFDFDSYSVKDEYQPLLQQHAQYLKS 98
    HPQRHVLIQGNTDERGTSEYNLALGQKRAEAVRRALALLGVADSQMEAV
    SLGKEKPLATGHDEASWAQNRRADLVYQQ
    B. AMBIFARIA LDDKANAGAVSTQPSADNVAQVNVDPLNDPNSPLAKRSIYFDFDSYSVKDEYQPL+QQHAQYLKSHP 96
    QRHVLIQGNTDERGTSEYNLALGQKRAEAVRRA+ALLGV DSQMEAVS
    LGKEKPLA+GHDEASWAQNRRADLVYQQ
    B. VIETNAMIENSIS LDDKAN-AGAVSTQPSADNVAQVNVDPLNDPNSPLAKRSIYFDFDSYSVKDEYQPLLQQHAQYLKSH 97
    PQRHVLIQGNTDERGTSEYNLALGQKRAEAVRRA+ALLGV DSQMEAVS
    LGKEKPLATGHDEASWAQNRRADLVYQQ
    B. UBONENSIS LD+KANAGA+STQP++DNVAQV VDPLNDPNSPLAKRSIYFDFDSYSVKDEYQPL+ 92
    QQHAQYLKSHPQRHVLIQGNTDERGTSEYNLALGQKRAEAVRRA+ALLGVADSQMEAYSLGKEKPLA
    GHDEASWAQNRR+DLVYQQ
    B.THAILANDENSIS LD+AN-GAVSTQP++NAQV VDPLNDPNSPLAKRSIYFDFDSYSV+D+YQPLLQ 88
    QHAQYLKSHPQRH+LIQGNTDERGTSEYNLALGQKRAEAVRRAL+LLGV DSQMEAVSLGK
    EKPLATGHDEASWAQNRRADLVYQQ
    B. GRAMINIS LD+AN-GAVSTQP+++VAQVNVDPLNDPNSPLAKRSIYFDFDSYSVKD+ 88
    YQPLLQQH+QYLKSHPQRHVLIQGNTDERGTSEYNLALGQKRAEAVRR+L+L+
    GV DSQMEAVSLGKEKPLATGHDE+SWAQNRRADLVYQQ
    B. OKLAHOMENSIS LD+ AN-GAVSTQP++NVAQV VDPLNDPNSPLAKRSIYFDFDSYSV+D+ 87
    YQPLLQQHAQYLK HPQRH+LIQGNTDERGTSEYNLALGQKRAEAVRRAL+LLGV
    DSQMEAVSLGKEKPLA GHDEASWAQNRRADLVYQQ
    B. PSEUDOMALLEI LD+ AN G-AVSTQP++NVAQV VDPLNDPNSPLAKRSIYFDFDSYSV+D+ 87
    YQPLLQQHAQYLKSHPQRH+LIQGNTDERGTSEYNLALGQKRAEAVRRAL+LLGV D+
    QMEAVSLGKEKPLA GHDEASWAQNRRADLVYQQ
    B. XENOVARANS LD+ AN G-+VS QP+ ++VA V VDPLNDPNSPLAKRSIYFDFDSYSVKD+ 86
    YQ LLQQHAQYLKSHPQRH+LIQGNTDERGTSEYNLALGQKRAEAVRR+L+L+GV
    DSQMEAVSLGKEKP ATGHDE+SWAQNRRADLVYQQ
    B. PHYTOFIRMANS LD+ AN-G VS QP+ ++VAVNVDPLNDPNSPLAKRSIYFDFDSYSVKD+YQ 86
    LLQQHAQYLKSHPQRHVLIQGNTDERGTSEYNLALGQKRAEAVRR+L+L+GV
    DSQMEAVSLGKEKP ATGHDE+SWAQNRRADLVYQQ
    B. PHYMATUM LD+ A- GA QP+ ++VA VNVDPLNDPNSPLAKRSIYFDFDSYSVKD+ 86
    YQPLLQQHAQYLKSHPQRHVLIQGNTDERGTSEYNLALGQKRAEAVRR+L+LLGV DS+
    MEAVSLGKEKP ATGHDEASWAQNRRADLVYQQ
    B. METALLIDURANS LDD++GA- A NVA V+V-DPLNDPN PLAKRSIYFDFDSYSVK +YQ + 73
    LQ H+QYL S+ R +LIQGNTDERGTSEYNLALGQKRAEAVRR+LA +GV
    DSQMEAVSLGKEKP ATGHD+ASWA+NRRAD+VY
    B. EUTROPHA LDD ANAGA AD-V V+V-DPLNDPNSPLAKRSIYFDFDSYSVK EYQ+L +HA+YL 73
    S+ R +LIQGNTDERGTSEYNLALGQKRAEAVRR+LA+GV+
    DSQMEAVSLGKEKP+TGHDEA+WA+NRRAD+Y
    B. SOLANACEARUM LDD-K G +T NV V+V-DL DPNSPLAKRSIYFDFDSY+VK EYQ LL 74
    QHA+YL+SH QR VLIQGNTDERGTSEYNLALGQKRAEAVRRAL+ GV
    DSQMEAVSLGKEKP ATGHDE SWAQNRR+D+VY

    Intranasal Immunization with OMP-NE Induces Anti-OMP Specific Antibodies
  • The humoral immune responses against the OMP induced by nasal vaccine with OMP were characterized in vivo in the CD-1 mice. Intranasal vaccination with either 5 μg or 15 μg OMP preparations+NE (OMP-NE) resulted in high serum titers of OMP-specific IgG antibodies of 2.8×105 and 5.1×105 at 6 weeks, respectively, following primary vaccination (See FIG. 30A). OMPs without adjuvant were immunogenic and resulted in serum anti-OMP IgG titers of 1.9×104 and 3.8×104 six weeks following primary i.n. immunization. However, treatment groups with NE as an adjuvant responded significantly higher (13 to 30 fold) than groups without adjuvant at all time points following the boost (p<0.05). Further, mice immunized with OMP in PBS did not demonstrate a significant boost effect following the second vaccination (See FIG. 30A).
  • To further characterize the OMP-NE vaccine, the OMP-NE composition's ability to elicit specific antibody production in bronchiolar secretions was evaluated. Mucosal antibody production may be important for protection against Burkholderia colonization since secretory antibodies are thought to be critical effectors in protection against mucosal respiratory pathogens (See, e.g., Nelson et al., 1993, J Med Microbiol 39, 39-47). Mucosal immune responses were evaluated in bronchoalveolar lavage (BAL) fluid of animals immunized with 5 μg OMP-NE or 5 μg OMP in PBS. Comparable levels of anti-OMP antibodies in BALs were detected in mice immunized either with OMP-NE or OMP without NE adjuvant (See FIG. 30B).
  • OMP-NE Immunization Yields a Balanced Th1/Th2 Cellular Response
  • The analysis of the serum IgG subclass was performed to determine the T helper-type bias of the cellular response. The pattern of IgG subclass distribution shows that i.n. immunization with OMP-NE produced increased levels of Th1-type IgG2b versus Th2-type IgG1 subclass antibodies (p=0.048) (See FIG. 31A). In comparison, immunization with OMP in PBS produced similar levels of IgG1 and IgG2b antibodies (See FIG. 31A).
  • OMP-specific cellular responses were characterized in splenocytes obtained 6 weeks following primary vaccination with OMP-NE. The cells were stimulated with endotoxin-depleted OMP and then evaluated for specific cytokine production. In splenocytes collected from OMP-NE vaccinated mice, Th1 cytokines IFN-γ and IL-2 production increased 22.1 and 18.6-fold respectively over that of splenocytes from non-vaccinated mice (p=0.01 and 0.003, respectively) (See FIG. 31B). IFN-γ was equally induced in splenocytes from mice immunized with OMP in PBS. The Th2 cytokines IL-4, IL-5, IL-6, and IL-10 were minimally induced (≦5.22-fold increase) in both OMP-NE and OMP immunized mice in equivalent amounts. The pattern of splenic cytokine expression and the IgG subclass distribution results indicated a balanced Th1/Th2 response to OMP-NE vaccination.
  • Antibody Response is Directed Toward OMP Lipoprotein and LPS
  • It has been previously documented that LPS is intrinsically associated with Burkholderia spp. OMP preparations (See, e.g., Bertot et al., 2007, Infect Immun 75, 2740-2752). To investigate the specificity of immune response and to evaluate the possible immune enhancing role of endotoxin in B. cencocepacia OMP preparations, serum from mice immunized with OMP-NE was analyzed using ELISA with either crude OMP, endotoxin-depleted OMP, or LPS. The highest IgG reactivity was against the endotoxin-depleted and crude OMP preparations. The statistical analysis indicated a significantly higher response in endotoxin-depleted OMP versus LPS (OD405=2.37±0.11 (p=0.001)) and crude OMP versus LPS (OD405=1.49±0.14 (p=0.01)). However, the serum also contained significant levels of IgG anti-LPS antibodies (OD405=0.59±0.05).
  • To clarify the role of endotoxin in the OMP preparation, a proteolytic digest of the crude OMP and endotoxin-depleted OMP preparations were performed with proteinase K. The immunogenic epitopes were determined by western blot analysis comparing serum antibodies generated in mice vaccinated with OMP in PBS versus from mice vaccinated with OMP-NE (See FIG. 32). The results clearly indicated that antibody response was directed mainly toward 62 KDa and 17 KDa proteins contained within the OMP preparation. This was confirmed by the complete absence of the protein bands in the proteolytically digested OMP preparations (See FIG. 32 OMP-NE serum western blot). Further, when the OMP preparations were probed with commercial monoclonal anti-LPS antibody, LPS was detected as an intrinsic part of the OMP preparation. However, the proteolytic digest diminished the LPS detection but not totally. Overall, the present invention provides that nasal immunization with OMP-NE enhances the production of antibodies specific against OMP proteins (See FIG. 32).
  • Intranasal Vaccination with OMP-NE Results in Cross-Protective Neutralizing Serum Antibody
  • B. cenocepacia neutralizing antibodies were determined using a colony reduction assay. The mice intranasally vaccinated with 5 μg or 15 μg OMP-NE produced serum-neutralizing antibodies that inhibited 92.5% or 98.3% of B. cenocepacia colonies, respectively, as compared to serum from non-vaccinated mice (See FIG. 33). The mice vaccinated with OMP lacking adjuvant also demonstrated a relatively high level of inhibition, resulting in 33.3% and 46.7% (for 5 μg and 15 μg OMP-PBS, respectively) cfu reduction. The statistical analysis showed that B. cenocepacia growth was significantly inhibited by serum derived from OMP-NE as compared to serum from those who were immunized with the OMP preparation alone (See FIG. 33).
  • To evaluate if the OMP-NE can produce cross-reactive protection from other bacterial strains, serum from mice vaccinated with 15 μg OMP-NE was also incubated with B. multivorans. The B. multivorans was selected because it is the most common isolate cultured from CF patients infected with Bcc organisms (See, e.g., Baldwin et al., 2008, J Clin Microbiol 46, 290-295). The serum inhibited B. multivorans growth by 80.1% and by 49.8% derived from mice vaccinated with OMP-NE and OMP without adjuvant, respectively, suggesting significant cross-protective antibodies following immunization with either OMP-NE or OMP in PBS (See FIG. 33).
  • Immunization with OMP-NE Protects Against Pulmonary B. cenocepacia Challenge and Reduces Incidence of Sepsis
  • The protective effect of intranasal immunization was further tested in vivo in a lung infection model. The clearance of B. cenocepacia from pulmonary tissue was evaluated 6 days following intra-tracheal inoculation in mice that were intranasally immunized with the OMP-NE or the OMP without an adjuvant. Vaccination with 15 μg OMP-NE resulted in significantly higher rates of pulmonary clearance (p=9.2×10−3) as compared to the non-vaccinated mice (See FIG. 34A). At day 6, the average pulmonary bacterial load was 22.5±26.2 cfu in mice vaccinated with 15 μg OMP-NE, compared to 1.28×106±3.36×106 cfu per lung in non-vaccinated mice, representing a greater than 5-log reduction in the bacterial load.
  • Splenic colonization following pulmonary inoculation with B. cenocepacia was evaluated as a means of assessing sepsis (See FIG. 34B). Vaccination with 15 μg OMP-NE significantly reduced the incidence of bacteria in the spleens 6 days following the pulmonary challenge of individual mice from an average of 3.54×103±6.97×103 cfu per spleen in non-vaccinated mice to 2.5±5 cfu per spleen in vaccinates (p=0.0307).
  • Intranasal Immunization with OMP-NE Attenuates Systemic Illness after B. cenocepacia Infection
  • B. cenocepacia infection resulted in greater loss of thermoregulation by day 6 in the non-vaccinated mice than in the mice immunized with OMP-NE or OMP without the adjuvant. The mean body temperature 6 days following infection with B. cenocepacia was significantly lower in non-vaccinated mice (35.4° C.±0.7) as compared to mice vaccinated with 15 μg OMP-NE (36.8° C.±0.31, p=0.008), 5 μg OMP-NE (37.1° C.±0.42, p=0.008), or 5 μg OMP without the adjuvant (36.8° C.±0.61, p=0.01).
  • The total peripheral leukocyte counts determined at the time of sacrifice in the non-vaccinated mice were significantly higher from the values observed for the mice vaccinated with 15 μg OMP-NE (p=0.047), 5 μg OMP-NE (p=0.026), or 15 μg OMP in PBS (p=0.032). The ratio of polymorphonuclear leukocytes to total peripheral leukocytes were also significantly higher in non-vaccinated mice (76.0%) compared to mice vaccinated with 15 μg OMP-NE (47.7%, p=0.02), 5 μg OMP-NE (45.6%, p=0.024), or 15 μg OMP in PBS (48.7%, p=0.04) (See Table 26, below). In combination with the splenic colonization results described above (See FIG. 39B), the present invention provides that immunization with OMP-NE resulted in significantly decreased systemic disease following infection with B. cenocepacia.
  • TABLE 26
    Ratio of polymorphonuclear leukocytes to total peripheral leukocytes.
    Mean Polymorphonuclear Leukocytes
    Immunization Concentration % Total Peripheral
    Group (109/L) Leukocyte Count
     5 μg OMP-NE 1.99 ± 0.20* 45.6*
     5 μg OMP in PBS 3.23 ± 0.29 59.7
    15 μg OMP-NE 2.58 ± 0.48* 47.7*
    15 μg OMP in PBS 2.56 ± 0.55* 48.7*
    Non-Vaccinated 7.29 ± 1.35 76.0
  • All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described compositions and methods of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the relevant fields are intended to be within the scope of the present invention.

Claims (34)

1. A method of treating or preventing a respiratory infection in a subject having Cystic fibrosis (CF), wherein:
(a) the method comprises administering a nanoemulsion to the subject;
(b) the subject is susceptible to or has an infection by one or more gram-positive or gram-negative bacterial species;
(c) the nanoemulsion comprises:
(i) water;
(ii) at least one organic solvent;
(iii) at least one surfactant; and
(iv) at least one oil; and
(d) wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
2. A method of treating or preventing an infection in a subject having a burn wound, wherein:
(a) the method comprises administering a nanoemulsion to the subject;
(b) the subject is susceptible to or has an infection by one or more gram-positive or gram-negative bacterial species;
(c) the nanoemulsion comprises:
(i) water;
(ii) at least one organic solvent;
(iii) at least one surfactant; and
(iv) at least one oil; and
(d) wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
3. A method of treating or preventing an Haemophilus influenzae infection in a subject wherein:
(a) the method comprises administering a nanoemulsion to the subject having or at risk of having a Haemophilus influenzae infection;
(b) the nanoemulsion comprises:
(i) water;
(ii) at least one organic solvent;
(iii) at least one surfactant; and
(iv) at least one oil; and
(c) wherein the nanoemulsion comprises droplets having an average diameter of less than about 1000 nm.
4. The method of claim 1, wherein the respiratory infection is associated with a bacterial biofilm present in the lungs of the subject.
5. The method of claim 1, further comprising administering one or more antibiotics either before, during, or after administration of the nanoemulsion.
6. The method of claim 5, wherein the administration of a nanoemulsion and at least one antibiotic is synergistic as defined by a fractional inhibitory concentration (FIC) index, a fractional bactericidal concentration (FBC) index, or a combination thereof.
7. The method of claim 6, wherein the antibiotic is any antibiotic, for example a polymyxin antibiotic (e.g. colistin) or any aminoglycoside (e.g. tobramycin).
8. The method of claim 5, wherein the nanoemulsion does not exhibit any antagonism with the antibiotic.
9. The method of claim 1, wherein the bacterial species is selected from the group consisting of Staphylococcus spp., Haemophilus spp., Pseudomonas spp., Burkholderia spp., Acinetobacter spp, Stenotrophomonas spp., Escherichia spp., Klebsiella spp., and Proteus spp.
10. The method of claim 9, wherein the subject is susceptible to or has an infection by one or more bacterial species selected from the group consisting of Pseudomonas aeruginosa, B. cenocepacia, A. baumannii, Stenotrophomonas maltophilia, Staphylococcus aureus, H. influenzae, E. coli, K. pneumoniae, and Proteus mirabilis.
11. The method of claim 1, wherein the nanoemulsion exhibits minimal or no toxicity or side effects.
12. The method of claim 1, wherein the minimum inhibitory concentration (MIC), the minimum bactericidal concentration (MBC), or a combination thereof for the nanoemulsion demonstrate bacteriostatic or bactericidal activity for the nanoemulsion.
13. The method of claim 1, wherein one or more bacterial species may exhibit resistance against one or more antibiotics.
14. The method of claim 1, wherein the bacterial species is methicillin-resistant Staphylococcus aureus (MRSA).
15. The method of claim 1, wherein the nanoemulsion does not exhibit resistance.
16. The method of claim 1, wherein the nanoemulsion comprises:
(a) water;
(b) ethanol or glycerol;
(c) cetylpyridinium chloride (CPC), or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824);
(d) soybean oil; and
(e) Poloxamer 407, Tween 80, or Tween 20.
17. The method of claim 16, wherein the nanoemulsion further comprises EDTA.
18. The method of claim 1, wherein the nanoemulsion is delivered via inhalation, topically to a mucosal surface, via nebulization, or via any combination thereof.
19. The method of claim 2, wherein the nanoemulsion is delivered topically, topically to a mucosal surface, via nebulization, or via any combination thereof.
20. The method of claim 3, wherein the nanoemulsion is delivered via inhalation, topically to a mucosal surfaces, via nebulization, or via any combination thereof.
21. The method of claim 1, wherein:
(a) the nanoemulsion droplets have an average diameter selected from the group consisting of less than about 950 nm, less than about 900 nm, less than about 850 nm, less than about 800 nm, less than about 750 nm, less than about 700 nm, less than about 650 nm, less than about 600 nm, less than about 550 nm, less than about 500 nm, less than about 450 nm, less than about 400 nm, less than about 350 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 100 nm, greater than about 50 nm, greater than about 70 nm, greater than about 125 nm, and any combination thereof;
(b) the nanoemulsion droplets have an average diameter greater than about 125 nm and less than about 300 nm;
(c) the nanoemulsion droplets have an average diameter of from about 300 nm to about 600 nm; or
(d) the nanoemulsion droplets have an average diameter of from about 150 nm to about 400 nm.
22. The method of claim 1, wherein the nanoemulsion comprises:
(a) an aqueous phase;
(b) about 1% oil to about 80% oil, or about 5% oil to about 80% oil;
(c) about 0.1% organic solvent to about 50% organic solvent, or about 0.1% organic solvent to about 10% organic solvent;
(d) at least one surfactant present in an amount of about 0.001% surfactant to about 10% surfactant;
(e) at least one non-ionic surfactant present in an amount of about 0.1% to about 10%;
(f) at least one cationic agent present in an amount of about 0.01% to about 3%;
(g) about 0.0005% to about 1% of a chelating agent; or
(h) any combination thereof.
23. The method of claim 1, wherein:
(a) the nanoemulsion has a narrow range of MIC (minimum inhibitory concentration) and MBC (minimum bactericidal concentrations) values;
(b) the MIC and MBC for the nanoemulsion differ by less than or equal to four-fold, meaning that the nanoemulsion is bactericidal;
(c) the MIC and MBC for the nanoemulsion differ by greater than four-fold, meaning that the nanoemulsion is bacteriostatic;
(d) any combination thereof.
24. The method of claim 1, wherein the nanoemulsion is stable:
(a) at about 40° C. and about 75% relative humidity for a time period selected from the group consisting of up to about 1 month, up to about 3 months, up to about 6 months, up to about 12 months, up to about 18 months, up to about 2 years, up to about 2.5 years, and up to about 3 years;
(b) at about 25° C. and about 60% relative humidity for a time period selected from the group consisting of up to about 1 month, up to about 3 months, up to about 6 months, up to about 12 months, up to about 18 months, up to about 2 years, up to about 2.5 years, up to about 3 years, up to about 3.5 years, up to about 4 years, up to about 4.5 years, and up to about 5 years;
(c) at about 4° C. for a time period selected from the group consisting of up to about 1 month, up to about 3 months, up to about 6 months, up to about 12 months, up to about 18 months, up to about 2 years, up to about 2.5 years, up to about 3 years, up to about 3.5 years, up to about 4 years, up to about 4.5 years, up to about 5 years, up to about 5.5 years, up to about 6 years, up to about 6.5 years, and up to about 7 years; or
(d) any combination thereof.
25. The method of claim 1, wherein the organic solvent:
(a) is selected from the group consisting of a C1-C12 alcohol, diol, triol, dialkyl phosphate, tri-alkyl phosphate, and combinations thereof;
(b) is selected from the group consisting of a nonpolar solvent, a polar solvent, a protic solvent, an aprotic solvent, semi-synthetic derivatives thereof, and combinations thereof;
(c) is selected from the group consisting of tri-n-butyl phosphate, ethanol, methanol, isopropyl alcohol, glycerol, medium chain triglycerides, diethyl ether, ethyl acetate, acetone, dimethyl sulfoxide (DMSO), acetic acid, n-butanol, butylene glycol, perfumers alcohols, isopropanol, n-propanol, formic acid, propylene glycols, glycerol, sorbitol, industrial methylated spirit, triacetin, hexane, benzene, toluene, diethyl ether, chloroform, 1,4-dixoane, tetrahydrofuran, dichloromethane, acetone, acetonitrile, dimethylformamide, dimethyl sulfoxide, formic acid, semi-synthetic derivatives thereof, and any combination thereof; and
(d) any combination thereof.
26. The method of claim 1, wherein the oil is:
(a) any cosmetically or pharmaceutically acceptable oil;
(b) non-volatile;
(c) selected from the group consisting of animal oil, vegetable oil, natural oil, synthetic oil, hydrocarbon oils, silicone oils, and semi-synthetic derivatives thereof;
(d) selected from the group consisting of mineral oil, squalene oil, flavor oils, silicon oil, essential oils, water insoluble vitamins, Isopropyl stearate, Butyl stearate, Octyl palmitate, Cetyl palmitate, Tridecyl behenate, Diisopropyl adipate, Dioctyl sebacate, Menthyl anthranhilate, Cetyl octanoate, Octyl salicylate, Isopropyl myristate, neopentyl glycol dicarpate cetols, Ceraphyls®, Decyl oleate, diisopropyl adipate, C12-15 alkyl lactates, Cetyl lactate, Lauryl lactate, Isostearyl neopentanoate, Myristyl lactate, Isocetyl stearoyl stearate, Octyldodecyl stearoyl stearate, Hydrocarbon oils, Isoparaffin, Fluid paraffins, Isododecane, Petrolatum, Argan oil, Canola oil, Chile oil, Coconut oil, corn oil, Cottonseed oil, Flaxseed oil, Grape seed oil, Mustard oil, Olive oil, Palm oil, Palm kernel oil, Peanut oil, Pine seed oil, Poppy seed oil, Pumpkin seed oil, Rice bran oil, Safflower oil, Tea oil, Truffle oil, Vegetable oil, Apricot (kernel) oil, Jojoba oil (simmondsia chinensis seed oil), Grapeseed oil, Macadamia oil, Wheat germ oil, Almond oil, Rapeseed oil, Gourd oil, Soybean oil, Sesame oil, Hazelnut oil, Maize oil, Sunflower oil, Hemp oil, Bois oil, Kuki nut oil, Avocado oil, Walnut oil, Fish oil, berry oil, allspice oil, juniper oil, seed oil, almond seed oil, anise seed oil, celery seed oil, cumin seed oil, nutmeg seed oil, leaf oil, basil leaf oil, bay leaf oil, cinnamon leaf oil, common sage leaf oil, eucalyptus leaf oil, lemon grass leaf oil, melaleuca leaf oil, oregano leaf oil, patchouli leaf oil, peppermint leaf oil, pine needle oil, rosemary leaf oil, spearmint leaf oil, tea tree leaf oil, thyme leaf oil, wintergreen leaf oil, flower oil, chamomile oil, clary sage oil, clove oil, geranium flower oil, hyssop flower oil, jasmine flower oil, lavender flower oil, manuka flower oil, Marhoram flower oil, orange flower oil, rose flower oil, ylang-ylang flower oil, Bark oil, cassia Bark oil, cinnamon bark oil, sassafras Bark oil, Wood oil, camphor wood oil, cedar wood oil, rosewood oil, sandalwood oil), rhizome (ginger) wood oil, resin oil, frankincense oil, myrrh oil, peel oil, bergamot peel oil, grapefruit peel oil, lemon peel oil, lime peel oil, orange peel oil, tangerine peel oil, root oil, valerian oil, Oleic acid, Linoleic acid, Oleyl alcohol, Isostearyl alcohol, semi-synthetic derivatives thereof, and combinations thereof; or
(d) any combination thereof.
27. The method of claim 1, wherein the nanoemulsion comprises a volatile oil and wherein:
(a) the volatile oil is the organic solvent;
(b) the volatile oil is present in addition to an organic solvent;
(c) the volatile oil is a terpene, monoterpene, sesquiterpene, carminative, azulene, semi-synthetic derivatives thereof, or combinations thereof;
(d) the volatile oil is selected from the group consisting of a terpene, monoterpene, sesquiterpene, carminative, azulene, menthol, camphor, thujone, thymol, nerol, linalool, limonene, geraniol, perillyl alcohol, nerolidol, farnesol, ylangene, bisabolol, farnesene, ascaridole, chenopodium oil, citronellal, citral, citronellol, chamazulene, yarrow, guaiazulene, chamomile, semi-synthetic derivatives thereof, and combinations thereof; or
(e) the nanoemulsion comprises a silicone component and the volatile oil present in the silicone component is different than the oil in the oil phase;
(f) the nanoemulsion comprises a silicone component and the silicone component comprises at least one volatile silicone oil, wherein the volatile silicone oil can be the sole oil in the silicone component or it can be combined with other silicone and non-silicone oils, and wherein the other oils can be volatile or non-volatile;
(g) the nanoemulsion comprises a silicone component and the silicone component is selected from the group consisting of methylphenylpolysiloxane, simethicone, dimethicone, phenyltrimethicone (or an organomodified version thereof), alkylated derivatives of polymeric silicones, cetyl dimethicone, lauryl trimethicone, hydroxylated derivatives of polymeric silicones, such as dimethiconol, volatile silicone oils, cyclic and linear silicones, cyclomethicone, derivatives of cyclomethicone, hexamethylcyclotrisiloxane, octamethylcyclotetrasiloxane, decamethylcyclopentasiloxane, volatile linear dimethylpolysiloxanes, isohexadecane, isoeicosane, isotetracosane, polyisobutene, isooctane, isododecane, semi-synthetic derivatives thereof, and combinations thereof; or
(h) a combination thereof.
28. The method of claim 1 further comprising:
(a) a chelating agent;
(b) at least one preservative;
(c) at least one pH adjuster;
(d) at least one buffer;
(e) at least one antibiotic agent; or
(f) any combination thereof.
29. The method of claim 28, wherein:
(a) the chelating agent (i) is present in an amount of about 0.0005% to about 1%; (ii) the chelating agent is selected from the group consisting of ethylenediamine, ethylenediaminetetraacetic acid, and dimercaprol; (iii) the chelating agent is ethylenediaminetetraacetic acid; or (iv) any combination thereof;
(b) the preservative is selected from the group consisting of cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha-tocophernol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, sodium ascorbate, Bis(p-chlorophenyldiguanido) hexane, 3-(-4-chloropheoxy)-propane-1,2-diol, Methyl and methylchloroisothiazolinone, sodium metabisulphite, citric acid, edetic acid, chlorphenesin (3-(-4-chloropheoxy)-propane-1,2-diol), Kathon CG (methyl and methylchloroisothiazolinone), parabens (methyl, ethyl, propyl, butyl hydrobenzoates), phenoxyethanol (2-phenoxyethanol), Phenonip (phenoxyethanol, methyl, ethyl, butyl, propyl parabens), Phenoroc (phenoxyethanol 0.73%, methyl paraben 0.2%, propyl paraben 0.07%), Liquipar Oil (isopropyl, isobutyl, butylparabens), Liquipar PE (70% phenoxyethanol, 30% liquipar oil), Nipaguard MPA (benzyl alcohol (70%), methyl & propyl parabens), Nipaguard MPS (propylene glycol, methyl & propyl parabens), Nipasept (methyl, ethyl and propyl parabens), Nipastat (methyl, butyl, ethyl and propyel parabens), Elestab 388 (phenoxyethanol in propylene glycol plus chlorphenesin and methylparaben), Killitol (7.5% chlorphenesin and 7.5% methyl parabens), semi-synthetic derivatives thereof, and combinations thereof;
(c) the pH adjuster is selected from the group consisting of diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof;
(d) the buffer is selected from the group consisting of 2-Amino-2-methyl-1,3-propanediol, 2-Amino-2-methyl-1-propanol, L-(+)-Tartaric acid, ACES, ADA, Acetic acid, Ammonium acetate solution, Ammonium bicarbonate, Ammonium citrate dibasic, Ammonium formate, Ammonium oxalate monohydrate, Ammonium phosphate dibasic, Ammonium phosphate monobasic, Ammonium sodium phosphate dibasic tetrahydrate, Ammonium sulfate solution, Ammonium tartrate dibasic, BES buffered saline, BES, BICINE, BIS-TRIS, Bicarbonate buffer solution, Boric acid, CAPS, CHES, Calcium acetate hydrate, Calcium carbonate, Calcium citrate tribasic tetrahydrate, Citrate Concentrated Solution, Citric acid, hydrous, Diethanolamine, EPPS, Ethylenediaminetetraacetic acid disodium salt dihydrate, Formic acid solution, Gly-Gly-Gly, Gly-Gly, Glycine, HEPES, Imidazole, Lipoprotein Refolding Buffer, Lithium acetate dihydrate, Lithium citrate tribasic tetrahydrate, MES hydrate, MES monohydrate, MES solution, MOPS, Magnesium acetate solution, Magnesium acetate tetrahydrate, Magnesium citrate tribasic nonahydrate, Magnesium formate solution, Magnesium phosphate dibasic trihydrate, Oxalic acid dihydrate, PIPES, Phosphate buffered saline, piperazine, Potassium D-tartrate monobasic, Potassium acetate, Potassium bicarbonate, Potassium carbonate, Potassium chloride, Potassium citrate monobasic, Potassium citrate tribasic solution, Potassium formate, Potassium oxalate monohydrate, Potassium phosphate dibasic, Potassium phosphate dibasic, for molecular biology, anhydrous, Potassium phosphate monobasic, Potassium phosphate monobasic, Potassium phosphate tribasic monohydrate, Potassium phthalate monobasic, Potassium sodium tartrate, Potassium sodium tartrate tetrahydrate, Potassium tetraborate tetrahydrate, Potassium tetraoxalate dihydrate, Propionic acid, STE buffer, STET buffer, Sodium 5,5-diethylbarbiturate, Sodium acetate, Sodium acetate trihydrate, Sodium bicarbonate, Sodium bitartrate monohydrate, Sodium carbonate decahydrate, Sodium carbonate, Sodium citrate monobasic, Sodium citrate tribasic dihydrate, Sodium formate solution, Sodium oxalate, Sodium phosphate dibasic dihydrate, Sodium phosphate dibasic dodecahydrate, Sodium phosphate dibasic solution, Sodium phosphate monobasic dihydrate, Sodium phosphate monobasic monohydrate, Sodium phosphate monobasic solution, Sodium pyrophosphate dibasic, Sodium pyrophosphate tetrabasic decahydrate, Sodium tartrate dibasic dihydrate, Sodium tartrate dibasic solution, Sodium tetraborate decahydrate, TAPS, TES, TM buffer solution, TNT buffer solution, TRIS Glycine buffer, TRIS acetate—EDTA buffer solution, TRIS buffered saline, TRIS glycine SDS buffer solution, TRIS phosphate EDTA buffer solution, Tricine, Triethanolamine, Triethylamine, Triethylammonium acetate buffer, Triethylammonium phosphate solution, Trimethylammonium acetate solution, Trimethylammonium phosphate solution, Tris-EDTA buffer solution, Trizma® acetate, Trizma® base, Trizma® carbonate, Trizma® hydrochloride, Trizma® maleate, or any combination thereof;
(e) the antibiotic agent is selected from the group consisting of Aminoglycosides, Ansamycins, Carbacephems, Carbapenems, Cephalosporins, Glycopeptides, Macrolides, Monobactams, Penicillins, Polypeptides, Polymyxins, Quinolones, Sulfonamides, Tetracyclines, and new classes of antibiotic agents; or
(f) any combination thereof.
30. The method of claim 1, wherein:
(a) the surfactant is selected from the group consisting of ethoxylated nonylphenol comprising 9 to 10 units of ethyleneglycol, ethoxylated undecanol comprising 8 units of ethyleneglycol, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate, polyoxyethylene (20) sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monostearate, sorbitan monooleate, ethoxylated hydrogenated ricin oils, sodium laurylsulfate, a diblock copolymer of ethyleneoxyde and propyleneoxyde, Ethylene Oxide-Propylene Oxide Block Copolymers, and tetra-functional block copolymers based on ethylene oxide and propylene oxide, Glyceryl monoesters, Glyceryl caprate, Glyceryl caprylate, Glyceryl cocate, Glyceryl erucate, Glyceryl hydroxysterate, Glyceryl isostearate, Glyceryl lanolate, Glyceryl laurate, Glyceryl linolate, Glyceryl myristate, Glyceryl oleate, Glyceryl PABA, Glyceryl palmitate, Glyceryl ricinoleate, Glyceryl stearate, Glyceryl thighlycolate, Glyceryl dilaurate, Glyceryl dioleate, Glyceryl dimyristate, Glyceryl disterate, Glyceryl sesuioleate, Glyceryl stearate lactate, Polyoxyethylene cetyl/stearyl ether, Polyoxyethylene cholesterol ether, Polyoxyethylene laurate or dilaurate, Polyoxyethylene stearate or distearate, polyoxyethylene fatty ethers, Polyoxyethylene lauryl ether, Polyoxyethylene stearyl ether, polyoxyethylene myristyl ether, a steroid, Cholesterol, Betasitosterol, Bisabolol, fatty acid esters of alcohols, isopropyl myristate, Aliphati-isopropyl n-butyrate, Isopropyl n-hexanoate, Isopropyl n-decanoate, Isoproppyl palmitate, Octyldodecyl myristate, alkoxylated alcohols, alkoxylated acids, alkoxylated amides, alkoxylated sugar derivatives, alkoxylated derivatives of natural oils and waxes, polyoxyethylene polyoxypropylene block copolymers, nonoxynol-14, PEG-8 laurate, PEG-6 Cocoamide, PEG-20 methylglucose sesquistearate, PEG40 lanolin, PEG-40 castor oil, PEG-40 hydrogenated castor oil, polyoxyethylene fatty ethers, glyceryl diesters, polyoxyethylene stearyl ether, polyoxyethylene myristyl ether, and polyoxyethylene lauryl ether, glyceryl dilaurate, glyceryl dimystate, glyceryl distearate, semi-synthetic derivatives thereof, and mixtures thereof;
(b) the surfactant is a non-ionic lipid selected from the group consisting of glyceryl laurate, glyceryl myristate, glyceryl dilaurate, glyceryl dimyristate, semi-synthetic derivatives thereof, and mixtures thereof;
(c) the surfactant is a polyoxyethylene fatty ether having a polyoxyethylene head group ranging from about 2 to about 100 groups;
(d) the surfactant is an alkoxylated alcohol having the structure shown in formula I below:

R5—(OCH2CH2)y—OH  Formula I
wherein R5 is a branched or unbranched alkyl group having from about 6 to about 22 carbon atoms and y is between about 4 and about 100, and preferably, between about 10 and about 100;
(e) the surfactant is an alkoxylated alcohol which is an ethoxylated derivative of lanolin alcohol;
(f) the surfactant is nonionic and is selected from the group consisting of nonoxynol-9, an ethoxylated surfactant, an alcohol ethoxylated, an alkyl phenol ethoxylated, a fatty acid ethoxylated, a monoalkaolamide ethoxylated, a sorbitan ester ethoxylated, a fatty amino ethoxylated, an ethylene oxide-propylene oxide copolymer, Bis(polyethylene glycol bis[imidazoyl carbonyl]), Brij®35, Brij® 56, Brij® 72, Brij® 76, Brij® 92V, Brij® 97, Brij® 58P, Cremophor® EL, Decaethylene glycol monododecyl ether, N-Decanoyl-N-methylglucamine, n-Decyl alpha-D-glucopyranoside, Decyl beta-D-maltopyranoside, n-Dodecanoyl-N-methylglucamide, n-Dodecyl alpha-D-maltoside, n-Dodecyl beta-D-maltoside, Heptaethylene glycol monodecyl ether, Heptaethylene glycol monotetradecyl ether, Heptaethylene glycol monododecyl ether, n-Hexadecyl beta-D-maltoside, Hexaethylene glycol monododecyl ether, Hexaethylene glycol monohexadecyl ether, Hexaethylene glycol monooctadecyl ether, Hexaethylene glycol monotetradecyl ether, Igepal CA-630, Methyl-6-O—(N-heptylcarbamoyl)-alpha-D-glucopyranoside, Nonaethylene glycol monododecyl ether, N-Nonanoyl-N-methylglucamine, Octaethylene glycol monodecyl ether, Octaethylene glycol monododecyl ether, Octaethylene glycol monohexadecyl ether, Octaethylene glycol monooctadecyl ether, Octaethylene glycol monotetradecyl ether, Octyl-beta-D-glucopyranoside, Pentaethylene glycol monodecyl ether, Pentaethylene glycol monododecyl ether, Pentaethylene glycol monohexadecyl ether, Pentaethylene glycol monohexyl ether, Pentaethylene glycol monooctadecyl ether, Pentaethylene glycol monooctyl ether, Polyethylene glycol diglycidyl ether, Polyethylene glycol ether W-1, Polyoxyethylene 10 tridecyl ether, Polyoxyethylene 100 stearate, Polyoxyethylene 20 isohexadecyl ether, Polyoxyethylene 20 oleyl ether, Polyoxyethylene 40 stearate, Polyoxyethylene 50 stearate, Polyoxyethylene 8 stearate, Polyoxyethylene bis(imidazolyl carbonyl), Polyoxyethylene 25 propylene glycol stearate, Saponin from Quillaja bark, Span® 20, Span® 40, Span® 60, Span® 65, Span® 80, Span® 85, Tergitol, Tergitol Type 15-S-12, Tergitol Type 15-S-30, Tergitol Type 15-S-5, Tergitol Type 15-S-7, Tergitol Type 15-S-9, Tergitol Type NP-10, Tergitol Type NP-4, Tergitol Type NP-40, Tergitol Type NP-7, Tergitol Type NP-9, Tergitol Type TMN-10, Tergitol Type TMN-6, Tetradecyl-beta-D-maltoside, Tetraethylene glycol monodecyl ether, Tetraethylene glycol monododecyl ether, Tetraethylene glycol monotetradecyl ether, Triethylene glycol monodecyl ether, Triethylene glycol monododecyl ether, Triethylene glycol monohexadecyl ether, Triethylene glycol monooctyl ether, Triethylene glycol monotetradecyl ether, Triton CF-21, Triton CF-32, Triton DF-12, Triton DF-16, Triton GR-5M, Triton QS-15, Triton QS-44, Triton X-100, Triton X-102, Triton X-15, Triton X-151, Triton X-200, Triton X-207, Triton X-114, Triton X-165, Triton X-305, Triton X-405, Triton X-45, Triton X-705-70, TWEEN® 20, TWEEN® 21, TWEEN® 40, TWEEN® 60, TWEEN® 61, TWEEN® 65, TWEEN® 80, TWEEN® 81, TWEEN® 85, Tyloxapol, n-Undecyl beta-D-glucopyranoside, Poloxamer 101, Poloxamer 105, Poloxamer 108, Poloxamer 122, Poloxamer 123, Poloxamer 124, Poloxamer 181, Poloxamer 182, Poloxamer 183, Poloxamer 184, Poloxamer 185, Poloxamer 188, Poloxamer 212, Poloxamer 215, Poloxamer 217, Poloxamer 231, Poloxamer 234, Poloxamer 235, Poloxamer 237, Poloxamer 238, Poloxamer 282, Poloxamer 284, Poloxamer 288, Poloxamer 331, Poloxamer 333, Poloxamer 334, Poloxamer 335, Poloxamer 338, Poloxamer 401, Poloxamer 402, Poloxamer 403, Poloxamer 407, Poloxamer 105 Benzoate, Poloxamer 182, Dibenzoate, semi-synthetic derivatives thereof, and combinations thereof;
(g) the surfactant is cationic and is selected from the group consisting of a quarternary ammonium compound, an alkyl trimethyl ammonium chloride compound, a dialkyl dimethyl ammonium chloride compound, Benzalkonium chloride, Benzyldimethylhexadecylammonium chloride, Benzyldimethyltetradecylammonium chloride, Benzyldodecyldimethylammonium bromide, Benzyltrimethylammonium tetrachloroiodate, Cetylpyridinium chloride, Dimethyldioctadecylammonium bromide, Dodecylethyldimethylammonium bromide, Dodecyltrimethylammonium bromide, Ethylhexadecyldimethylammonium bromide, Girard's reagent T, Hexadecyltrimethylammonium bromide, N,N′,N′-Polyoxyethylene(10)-N-tallow-1,3-diaminopropane, Thonzonium bromide, Trimethyl(tetradecyl)ammonium bromide, 1,3,5-Triazine-1,3,5(2H,4H,6H)-triethanol, 1-Decanaminium, N-decyl-N,N-dimethyl-, chloride, Didecyl dimethyl ammonium chloride, 2-(2-(p-(Diisobutyl)cresosxy)ethoxy)ethyl dimethyl benzyl ammonium chloride, 2-(2-(p-(Diisobutyl)phenoxy)ethoxy)ethyl dimethyl benzyl ammonium chloride, Alkyl 1 or 3 benzyl-1-(2-hydroxethyl)-2-imidazolinium chloride, Alkyl bis(2-hydroxyethyl)benzyl ammonium chloride, Alkyl demethyl benzyl ammonium chloride, Alkyl dimethyl 3,4-dichlorobenzyl ammonium chloride (100% C12), Alkyl dimethyl 3,4-dichlorobenzyl ammonium chloride (50% C14, 40% C12, 10% C16), Alkyl dimethyl 3,4-dichlorobenzyl ammonium chloride (55% C14, 23% C12, 20% C16), Alkyl dimethyl benzyl ammonium chloride, Alkyl dimethyl benzyl ammonium chloride (100% C14), Alkyl dimethyl benzyl ammonium chloride (100% C16), Alkyl dimethyl benzyl ammonium chloride (41% C14, 28% C12), Alkyl dimethyl benzyl ammonium chloride (47% C12, 18% C14), Alkyl dimethyl benzyl ammonium chloride (55% C16, 20% C14), Alkyl dimethyl benzyl ammonium chloride (58% C14, 28% C16), Alkyl dimethyl benzyl ammonium chloride (60% C14, 25% C12), Alkyl dimethyl benzyl ammonium chloride (61% C11, 23% C14), Alkyl dimethyl benzyl ammonium chloride (61% C12, 23% C14), Alkyl dimethyl benzyl ammonium chloride (65% C12, 25% C14), Alkyl dimethyl benzyl ammonium chloride (67% C12, 24% C14), Alkyl dimethyl benzyl ammonium chloride (67% C12, 25% C14), Alkyl dimethyl benzyl ammonium chloride (90% C14, 5% C12), Alkyl dimethyl benzyl ammonium chloride (93% C14, 4% C12), Alkyl dimethyl benzyl ammonium chloride (95% C16, 5% C18), Alkyl didecyl dimethyl ammonium chloride, Alkyl dimethyl benzyl ammonium chloride (C12-16), Alkyl dimethyl benzyl ammonium chloride (C12-18), dialkyl dimethyl benzyl ammonium chloride, Alkyl dimethyl dimethybenzyl ammonium chloride, Alkyl dimethyl ethyl ammonium bromide (90% C14, 5% C16, 5% C12), Alkyl dimethyl ethyl ammonium bromide (mixed alkyl and alkenyl groups as in the fatty acids of soybean oil), Alkyl dimethyl ethylbenzyl ammonium chloride, Alkyl dimethyl ethylbenzyl ammonium chloride (60% C14), Alkyl dimethyl isopropylbenzyl ammonium chloride (50% C12, 30% C14, 17% C16, 3% C18), Alkyl trimethyl ammonium chloride (58% C18, 40% C16, 1% C14, 1% C12), Alkyl trimethyl ammonium chloride (90% C18, 10% C16), Alkyldimethyl(ethylbenzyl) ammonium chloride (C12-18), Di-(C8-10)-alkyl dimethyl ammonium chlorides, Dialkyl dimethyl ammonium chloride, Dialkyl methyl benzyl ammonium chloride, Didecyl dimethyl ammonium chloride, Diisodecyl dimethyl ammonium chloride, Dioctyl dimethyl ammonium chloride, Dodecyl bis(2-hydroxyethyl) octyl hydrogen ammonium chloride, Dodecyl dimethyl benzyl ammonium chloride, Dodecylcarbamoyl methyl dimethyl benzyl ammonium chloride, Heptadecyl hydroxyethylimidazolinium chloride, Hexahydro-1,3,5-tris(2-hydroxyethyl)-s-triazine, Myristalkonium chloride (and) Quat RNIUM 14, N,N-Dimethyl-2-hydroxypropylammonium chloride polymer, n-Tetradecyl dimethyl benzyl ammonium chloride monohydrate, Octyl decyl dimethyl ammonium chloride, Octyl dodecyl dimethyl ammonium chloride, Octyphenoxyethoxyethyl dimethyl benzyl ammonium chloride, Oxydiethylenebis(alkyl dimethyl ammonium chloride), Trimethoxysily propyl dimethyl octadecyl ammonium chloride, Trimethoxysilyl quats, Trimethyl dodecylbenzyl ammonium chloride, semi-synthetic derivatives thereof, and combinations thereof;
(h) the surfactant is anionic and is selected from the group consisting of a carboxylate, a sulphate, a sulphonate, a phosphate, Chenodeoxycholic acid, Chenodeoxycholic acid sodium salt, Cholic acid, ox or sheep bile, Dehydrocholic acid, Deoxycholic acid, Deoxycholic acid methyl ester, Digitonin, Digitoxigenin, N,N-Dimethyldodecylamine N-oxide, Docusate sodium salt, Glycochenodeoxycholic acid sodium salt, Glycocholic acid hydrate, synthetic, Glycocholic acid sodium salt hydrate, synthetic, Glycodeoxycholic acid monohydrate, Glycodeoxycholic acid sodium salt, Glycolithocholic acid 3-sulfate disodium salt, Glycolithocholic acid ethyl ester, N-Lauroylsarcosine sodium salt, N-Lauroylsarcosine solution, Lithium dodecyl sulfate, Lugol solution, Niaproof 4, Type 4,1-Octanesulfonic acid sodium salt, Sodium 1-butanesulfonate, Sodium 1-decanesulfonate, Sodium 1-dodecanesulfonate, Sodium 1-heptanesulfonate anhydrous, Sodium 1-nonanesulfonate, Sodium 1-propanesulfonate monohydrate, Sodium 2-bromoethanesulfonate, Sodium cholate hydrate, Sodium choleate, Sodium deoxycholate, Sodium deoxycholate monohydrate, Sodium dodecyl sulfate, Sodium hexanesulfonate anhydrous, Sodium octyl sulfate, Sodium pentanesulfonate anhydrous, Sodium taurocholate, Taurochenodeoxycholic acid sodium salt, Taurodeoxycholic acid sodium salt monohydrate, Taurohyodeoxycholic acid sodium salt hydrate, Taurolithocholic acid 3-sulfate disodium salt, Tauroursodeoxycholic acid sodium salt, Trizma® dodecyl sulfate, Ursodeoxycholic acid, semi-synthetic derivatives thereof, and combinations thereof;
(i) the surfactant is zwitterionic and is selected from the group consisting of an N-alkyl betaine, lauryl amindo propyl dimethyl betaine, an alkyl dimethyl glycinate, an N-alkyl amino propionate, CHAPS (minimum 98%), CHAPSO (minimum 98%), 3-(Decyldimethylammonio)propanesulfonate inner salt, 3-(Dodecyldimethylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylmyristylammonio)propanesulfonate, 3-(N,N-Dimethyloctadecylammonio)propanesulfonate, 3-(N,N-Dimethyloctylammonio)propanesulfonate inner salt, 3-(N,N-Dimethylpalmitylammonio)propanesulfonate, semi-synthetic derivatives thereof, and combinations thereof;
(j) the surfactant is polymeric and the polymeric surfactant is selected from the group consisting of a graft copolymer of a poly(methyl methacrylate) backbone with at least one polyethylene oxide (PEO) side chain, polyhydroxystearic acid, an alkoxylated alkyl phenol formaldehyde condensate, a polyalkylene glycol modified polyester with fatty acid hydrophobes, a polyester, semi-synthetic derivatives thereof, and combinations thereof; or
(k) any combination thereof.
31. The method of claim 1, wherein the nanoemulsion:
(a) comprises at least one cationic surfactant;
(b) comprises a cationic surfactant which is either cetylpyridinium chloride, or benzalkonium chloride, or alkyl dimethyl benzyl ammonium chloride (BTC 824), or a combination thereof;
(c) comprises a cationic surfactant, and wherein the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001%;
(d) comprises a cationic surfactant, and wherein the concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1.5%, less than about 1.0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, less than about 0.10%, greater than about 0.001%, greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, and greater than about 0.010%; or
(e) any combination thereof.
32. The method of claim 1, wherein:
(a) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant;
(b) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant;
(c) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a polysorbate nonionic surfactant;
(d) the nanoemulsion comprises at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant, and the non-ionic surfactant is present in a concentration of about 0.05% to about 10%, about 0.05% to about 7.0%, about 0.1% to about 7%, or about 0.5% to about 4%;
(e) the nanoemulsion comprises at least one cationic surfactant and at least one a nonionic surfactant, wherein the cationic surfactant is present in a concentration of about 0.05% to about 3% or about 0.01% to about 3%; or
(f) any combination thereof.
33. The method of claim 1, wherein the water is present in Phosphate Buffered Saline (PBS).
34. The method of claim 1, wherein the nanoemulsion is not absorbed systemically in the human subject, or very little of the nanoemulsion is absorbed systemically in the human subject.
US12/567,470 2008-09-26 2009-09-25 Nanoemulsion therapeutic compositions and methods of using the same Abandoned US20100092526A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/567,470 US20100092526A1 (en) 2008-09-26 2009-09-25 Nanoemulsion therapeutic compositions and methods of using the same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10055908P 2008-09-26 2008-09-26
US12/567,470 US20100092526A1 (en) 2008-09-26 2009-09-25 Nanoemulsion therapeutic compositions and methods of using the same

Publications (1)

Publication Number Publication Date
US20100092526A1 true US20100092526A1 (en) 2010-04-15

Family

ID=41796583

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/567,470 Abandoned US20100092526A1 (en) 2008-09-26 2009-09-25 Nanoemulsion therapeutic compositions and methods of using the same
US12/567,571 Active 2030-08-01 US8962026B2 (en) 2008-09-26 2009-09-25 Nanoemulsion therapeutic compositions and methods of using the same
US14/628,886 Active US9259407B2 (en) 2008-09-26 2015-02-23 Nanoemulsion therapeutic compositions and methods of using the same
US15/041,173 Abandoned US20160158179A1 (en) 2008-09-26 2016-02-11 Nanoemulsion therapeutic compositions and methods of using the same

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/567,571 Active 2030-08-01 US8962026B2 (en) 2008-09-26 2009-09-25 Nanoemulsion therapeutic compositions and methods of using the same
US14/628,886 Active US9259407B2 (en) 2008-09-26 2015-02-23 Nanoemulsion therapeutic compositions and methods of using the same
US15/041,173 Abandoned US20160158179A1 (en) 2008-09-26 2016-02-11 Nanoemulsion therapeutic compositions and methods of using the same

Country Status (7)

Country Link
US (4) US20100092526A1 (en)
EP (1) EP2349209A2 (en)
JP (1) JP5722782B2 (en)
CN (1) CN102223876A (en)
AU (1) AU2009296458A1 (en)
CA (1) CA2738245A1 (en)
WO (2) WO2010036938A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100203139A1 (en) * 2008-09-26 2010-08-12 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
US20100278812A1 (en) * 2001-06-22 2010-11-04 Cpex Pharmaceuticals, Inc. Nasal immunization
WO2012003361A3 (en) * 2010-07-02 2012-04-12 The Regents Of The University Of Michigan Nanoemulsion vaccines
WO2012078482A1 (en) * 2010-12-10 2012-06-14 Merck Sharp & Dohme Corp. Novel formulations which mitigate agitation-induced aggregation of immunogenic compositions
US20140193461A1 (en) * 2009-05-14 2014-07-10 The Regents Of The University Of Michigan Streptococcus vaccine compositions and methods of using the same
US8785376B2 (en) 2010-12-20 2014-07-22 Virginia Tech Intellectual Properties Isolation, identification, and uses of antifungal compounds
WO2014151449A1 (en) * 2013-03-15 2014-09-25 Purdue Research Foundation Antimicrobial compositions and methods of use
WO2013096697A3 (en) * 2011-12-20 2015-06-18 University Of Utah Research Foundation Isolation, identification, and used of antifungal compounds
US20150216902A1 (en) * 2012-07-27 2015-08-06 Kassem Khal Fardoussi PHARMACEUTICAL COMPOSITION OF MILK FAT GLOBULES (MFGs) ZINC FREE
JP2016168058A (en) * 2010-10-28 2016-09-23 栄研化学株式会社 Disk test specimen for detecting drug-resistant bacterium
US20160368953A1 (en) * 2015-06-18 2016-12-22 The United States of America, as represented by the Secretary of Agirculture Bacteriocin with novel activity
WO2017017625A1 (en) * 2015-07-27 2017-02-02 Sun Pharmaceutical Industries Limited A stable topical pharmaceutical composition comprising nanonized silver sulfadiazine
US9561271B2 (en) 2011-09-09 2017-02-07 Nanobio Corporation Nanoemulsion respiratory syncytial virus (RSV) subunit vaccine
US9572774B2 (en) 2011-05-19 2017-02-21 Savara Inc. Dry powder vancomycin compositions and associated methods
US20170347661A1 (en) * 2011-04-06 2017-12-07 Convatec Technologies Inc. Composition comprising antimicrobial metal ions and a quaternary cationic surfactant
US20180008711A1 (en) * 2015-02-05 2018-01-11 Marc Selner Triple antibiotic cationic ointment
WO2018098376A1 (en) * 2016-11-22 2018-05-31 Elektrofi, Inc. Particles comprising a therapeutic or diagnostic agent and suspensions and methods of use thereof
EP3329936A1 (en) 2011-08-22 2018-06-06 Nanobio Corporation Herpes simplex virus nanoemulsion vaccine
WO2019055370A1 (en) * 2017-09-12 2019-03-21 Academic Pharmaceuticals, Inc. Metolazone emulsion formulation
CN110333216A (en) * 2018-12-29 2019-10-15 厦门市普识纳米科技有限公司 The detection method of xacin-series antibiotic in a kind of animal foodstuff
US10675243B2 (en) 2015-06-19 2020-06-09 Global Health Solutions Llc Petrolatum-based compositions comprising cationic biocides
US10729698B2 (en) * 2015-11-03 2020-08-04 Zoetis Services Llc Sol-gel polymer composites and uses thereof
WO2020240472A1 (en) * 2019-05-28 2020-12-03 Aybar Ecotechnologies Corp. Wide-spectrum antibacterial pharmaceutical formulations comprising lysozyme and methods of using the same
CN112569187A (en) * 2019-09-27 2021-03-30 武汉科福新药有限责任公司 Tacrolimus nano eye emulsion and preparation method thereof
NL2027383A (en) 2020-01-24 2021-09-01 Aim Immunotech Inc Methods, compositions, and vaccines for treating a virus infection
US20210330645A1 (en) * 2018-09-07 2021-10-28 Massey University Antibacterial combinations
WO2021262728A1 (en) * 2020-06-23 2021-12-30 Virogenomics Biodevelopment, Inc. Compositions and methods for viral detection
US11252960B2 (en) 2017-01-31 2022-02-22 Kimberly-Clark Worldwide, Inc. Antibacterial composition including benzoic acid ester and methods of inhibiting bacterial growth utilizing the same
CN114246839A (en) * 2020-09-22 2022-03-29 鲁南制药集团股份有限公司 Oral nanoemulsion enteric capsule and preparation method thereof
US11318149B2 (en) * 2018-09-27 2022-05-03 Board Of Trustees Of Michigan State University Compositions and methods for inhibiting biofilm-forming bacteria
CN115007119A (en) * 2021-03-03 2022-09-06 中国科学院大连化学物理研究所 Zwitterion-containing hydrophilic microsphere and preparation and application thereof
US11459376B2 (en) 2019-09-13 2022-10-04 Elektrofi, Inc. Compositions and methods for the delivery of therapeutic biologics for treatment of disease
US11717488B2 (en) 2019-01-31 2023-08-08 Elektrofi, Inc. Particle formation and morphology
US11795192B2 (en) 2017-03-10 2023-10-24 Lakewood Amedex, Inc. Antimicrobial compounds, compositions, and uses thereof

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102648003B (en) * 2009-10-09 2016-01-13 儿童医疗中心有限公司 The whole-cell vaccines of selective splitting
US20130267490A1 (en) * 2010-08-02 2013-10-10 Ranbaxy Laboratories Limited Topical pharmaceutical composition comprising nanonized silver sulfadiazine and chlorhexidine gluconate
US9433580B2 (en) 2011-07-21 2016-09-06 Sun Pharmaceutical Industries Limited Topical pharmaceutical composition comprising nanonized silver sulfadiazine
CN101947312B (en) * 2010-08-26 2012-08-15 董萍 Modified polyethylene glycol superoxide dismutase (mPEG-SOD) nanoemulsion capable of being directly externally used for skin and preparation method thereof
JP2014520805A (en) * 2011-07-06 2014-08-25 ナノバイオ コーポレーション Human respiratory syncytial virus vaccine
US9913862B2 (en) * 2011-10-26 2018-03-13 Trustees Of Boston University Methods of treating gram-negative microbial infections
CN102423315A (en) * 2011-11-23 2012-04-25 西北农林科技大学 Compound moxifloxacin nanoemulsion and preparation method thereof
CN102614452B (en) * 2012-04-25 2013-09-18 韩英 Chinese medicinal composition for effectively treating scleroderma
GB2503235A (en) * 2012-06-20 2013-12-25 Quest Vitamins Ltd Synergistic composition for minimising bacterial infection in animals
US20140377355A1 (en) * 2013-06-19 2014-12-25 Professional Compounding Centers Of America Levofloxacin Inhalation Composition
US9757209B2 (en) * 2013-07-03 2017-09-12 Essential Dental Systems, Inc. Compositions and methods for dental applications involving zinc-oxide cements
WO2015143386A1 (en) * 2014-03-21 2015-09-24 The Regents Of The University Of Michigan Topical nanoemulsion therapy for wounds
US20170014437A1 (en) * 2014-04-03 2017-01-19 Syracuse University Synthetic disugar hydrocarbons as natural analogs to control microbial behaviors
CN105338572B (en) * 2014-06-23 2020-07-31 北京三星通信技术研究有限公司 Data distribution method and device for split bearer in dual connectivity
ES2767294T3 (en) 2014-06-26 2020-06-17 Univ Rockefeller Acinetobacter lysines
WO2016048304A1 (en) * 2014-09-24 2016-03-31 Colgate-Palmolive Company Bioavailability of metal ions
EP4112076A1 (en) * 2014-10-10 2023-01-04 The Regents of The University of Michigan Nanoemulsion compositions for preventing, suppressing or eliminating allergic and inflammatory disease
CN104352566A (en) * 2014-10-21 2015-02-18 河南牧翔动物药业有限公司 Oil-in-water compound chlortetracycline nanoemulsion
CN104367690A (en) * 2014-10-21 2015-02-25 河南牧翔动物药业有限公司 Oil-in-water type compound lomefloxacin nano-emulsion
CN104274826B (en) * 2014-10-21 2016-04-20 河南牧翔动物药业有限公司 A kind of oil-in-water type compound colistin nano-emulsion
CN104352433A (en) * 2014-10-21 2015-02-18 河南牧翔动物药业有限公司 Oil-in-water compound antimicrobial cefazolin nanoemulsion medicine
CN104274499A (en) * 2014-10-21 2015-01-14 河南牧翔动物药业有限公司 Oil-in-water type compound minocyline antibacterial medicine
WO2016098009A1 (en) * 2014-12-16 2016-06-23 Dr. Reddy’S Laboratories Limited Lyophilized injectable compositions of daptomycin
WO2016145331A1 (en) * 2015-03-12 2016-09-15 Thermo Finnigan Llc Methods for data-dependent mass spectrometry of mixed biomolecular analytes
US9839652B2 (en) 2015-04-01 2017-12-12 Attostat, Inc. Nanoparticle compositions and methods for treating or preventing tissue infections and diseases
US11473202B2 (en) 2015-04-13 2022-10-18 Attostat, Inc. Anti-corrosion nanoparticle compositions
CN104856951A (en) * 2015-05-06 2015-08-26 华侨大学 Follicle stimulating hormone transdermal nanometer emulsion and preparation method thereof
KR101753842B1 (en) * 2015-05-12 2017-07-04 한국화학연구원 A sustained-release formulation comprising a fluoroquinolone antibiotics and a method for manufacturing the same
GB2556002B (en) 2015-07-07 2020-12-16 Bluewillow Biologics Inc Methods and compositions for nanoemulsion vaccine formulations
CN105055582B (en) * 2015-08-18 2018-09-04 云南同方科技有限公司 A kind of formula essential oil for the slight burn and scald of skin
WO2017035107A1 (en) 2015-08-24 2017-03-02 Smith & Nephew, Inc. Synergistic antibacterial activity of medium polarity oils in combination with antibacterial agents on bacterial biofilms
WO2017201390A1 (en) * 2016-05-19 2017-11-23 The Regents Of The University Of Michigan Novel adjuvant compositions
CN106076213A (en) * 2016-06-12 2016-11-09 上海应用技术学院 A kind of citral solid lipid nanoparticle under acid condition and preparation method thereof
US11246905B2 (en) 2016-08-15 2022-02-15 President And Fellows Of Harvard College Treating infections using IdsD from Proteus mirabilis
US10076551B2 (en) 2016-11-14 2018-09-18 FREDRICK COBBLE, Jr. Method for treating interstitial lung disease
CN106924730B (en) * 2017-02-14 2019-11-08 商丘美兰生物工程有限公司 Porcine pseudorabies virus Aloe Vera Gel nano emulsion adjuvant inactivated vaccine and preparation method thereof
CN106906688A (en) * 2017-03-03 2017-06-30 山东太阳生活用纸有限公司 A kind of Yankee dryer remover
JP6917078B2 (en) * 2017-03-10 2021-08-11 レイクウッド アメディックス、インコーポレイテッド Antibacterial compounds, compositions, and their use
BR112019021264B1 (en) * 2017-04-11 2023-10-24 Stepan Company METHOD FOR DISINFECTING AN INANIMATE SURFACE CONTAINING OR SUSPECTED OF CONTAINING BACTERIA CAUSING TUBERCULOSIS
JP7247164B2 (en) * 2017-07-17 2023-03-28 アンスティテュ ギュスタブ ルシ Pourable water-in-oil emulsion and its use
WO2019058338A1 (en) * 2017-09-24 2019-03-28 Aodh Lifesciences Private Limited Methods to produce liquid oxygen emulsion and administer intravascular liquid oxygen
CN107586792B (en) * 2017-09-27 2021-04-23 浙江海洋大学 Biological oil gelling agent and preparation method thereof
CN107496604A (en) * 2017-09-28 2017-12-22 贵州航生态农牧科技开发有限公司 A kind of camellia oil nanometer emulsion oral liquid and preparation method thereof
US20200345654A1 (en) * 2017-11-01 2020-11-05 Nihon Pharmaceutical Co., Ltd. Pharmaceutical composition, method for stabilizing pharmaceutical composition, and method for evaluating storage stability of pharmaceutical composition
US11646453B2 (en) 2017-11-28 2023-05-09 Attostat, Inc. Nanoparticle compositions and methods for enhancing lead-acid batteries
WO2019174277A1 (en) * 2018-03-13 2019-09-19 The Hong Kong Polytechnic University Phosphoethanolamine transferase inhibitors
US11324791B2 (en) 2018-04-02 2022-05-10 Imam Abdulrahman Bin Faisal University Antimicrobial extracts of Boswellia and thyme
CN112203683A (en) * 2018-04-10 2021-01-08 杜雷安教育基金会行政处 Burkholderia pseudorhizideus composite outer membrane vesicle as adjuvant
GB201809627D0 (en) * 2018-06-12 2018-07-25 Biorelevant Com Ltd Methods for preparing buffer solutions suitable for in vitro drug dissolution testing, drug solubility testing and/or drug profiling
CN110664920B (en) * 2018-07-03 2021-05-14 上海海洋大学 Medicine for resisting aquatic pathogenic stenotrophomonas maltophilia and preparation method thereof
CN110664901B (en) * 2018-07-03 2021-05-14 上海海洋大学 Medicine for resisting aquatic pathogenic bacillus mycoides and preparation method thereof
WO2020102494A1 (en) 2018-11-15 2020-05-22 Bluewillow Biologics, Inc. Nanoemulsion compositions having enhanced permeability
US20200155360A1 (en) * 2018-11-21 2020-05-21 Bocannaveda, Llc Nano-emulsified cannabinoid or oils in athletic tape and other products
PL428045A1 (en) * 2018-12-04 2020-06-15 Instytut Immunologii I Terapii Doświadczalnej Im. Ludwika Hirszfelda Polskiej Akademii Nauk Vaccine for the prevention and treatment of C. difficile infections and its application
CN109700694B (en) * 2019-01-26 2022-01-28 广东芭薇生物科技股份有限公司 Composition with moistening and moisturizing effects and application thereof
WO2020227415A1 (en) * 2019-05-06 2020-11-12 President And Fellows Of Harvard College Dna-degrading proteins and uses thereof
CN113840910A (en) * 2019-05-21 2021-12-24 合同会社予幸集团中央研究所 Method for washing extracellular vesicles
US20210007982A1 (en) * 2019-07-12 2021-01-14 Attostat, Inc. Use of nanoparticles for treating respiratory infections associated with cystic fibrosis
CN110257034A (en) * 2019-07-12 2019-09-20 中国石油化工股份有限公司 A kind of water mixing pipeline snaking de-plugging agent and its plug-removing device
CN114207101A (en) 2019-07-31 2022-03-18 埃科莱布美国股份有限公司 Deliming composition free of personal protective equipment
IT201900013983A1 (en) * 2019-08-05 2021-02-05 St Fisioterapici Ospitalieri Assay for the determination of susceptibility profiles to antibiotics in biofilm-producing microorganisms.
TR201917675A2 (en) * 2019-11-13 2021-05-21 Ulusal Bor Arastirma Enstituesue A WOUND CARE PRODUCT
JP7430381B2 (en) 2020-02-04 2024-02-13 水谷ペイント株式会社 Evaluation method for biofilm destruction ability
US20230113011A1 (en) * 2020-03-13 2023-04-13 Cmtx Biotech Inc. Methods of treating illnesses caused by coronavirus, swine flu and avian flu
KR102380976B1 (en) * 2020-05-14 2022-03-31 주식회사 아이코맥스 A skin cosmetic composition comprising goat's milk, hemp seed oil, manuca oil, Oenothera odorata oil and boswellia extract and the manufacuring method thereof
CN116437929A (en) * 2020-07-02 2023-07-14 普渡研究基金会 Inhalation formulations of antimicrobial compounds
US11052041B1 (en) * 2020-10-01 2021-07-06 King Abdulaziz University Nanotechnology-based nostril drops for relief of respiratory ailments
CN113521003B (en) * 2021-03-09 2022-06-28 山西利普达医药科技有限公司 Ophthalmic composition and preparation method and application thereof
WO2022271257A2 (en) * 2021-04-08 2022-12-29 West Vrginia University Cross-protective antigens for vaccination
CN117003898B (en) * 2023-09-27 2023-12-29 贝格探索(成都)科技有限公司 LHRH4-CRM197-LF multimeric recombinant protein and preparation method and application thereof

Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3854480A (en) * 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US4452775A (en) * 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
US4596556A (en) * 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4675189A (en) * 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
US4709017A (en) * 1985-06-07 1987-11-24 President And Fellows Of Harvard College Modified toxic vaccines
US4895452A (en) * 1988-03-03 1990-01-23 Micro-Pak, Inc. Method and apparatus for producing lipid vesicles
US4950740A (en) * 1987-03-17 1990-08-21 Cetus Corporation Recombinant diphtheria vaccines
US5057540A (en) * 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US5103497A (en) * 1989-11-14 1992-04-07 Hicks John W Flying spot endoscope
US5133974A (en) * 1989-05-05 1992-07-28 Kv Pharmaceutical Company Extended release pharmaceutical formulations
US5284656A (en) * 1991-03-15 1994-02-08 Amgen Inc. Pulmonary administration of granulocyte colony stimulating factor
US5407686A (en) * 1991-11-27 1995-04-18 Sidmak Laboratories, Inc. Sustained release composition for oral administration of active ingredient
US5451569A (en) * 1994-04-19 1995-09-19 Hong Kong University Of Science And Technology R & D Corporation Limited Pulmonary drug delivery system
US5565332A (en) * 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5592904A (en) * 1993-05-03 1997-01-14 Negre; Guy Method and devices for controlling the combustion of a four stroke engine
US5736152A (en) * 1995-10-27 1998-04-07 Atrix Laboratories, Inc. Non-polymeric sustained release delivery system
US5843711A (en) * 1992-05-06 1998-12-01 The Regents Of The University Of California Diphtheria toxin receptor-binding region
US5917017A (en) * 1994-06-08 1999-06-29 President And Fellows Of Harvard College Diphtheria toxin vaccines bearing a mutated R domain
US5985309A (en) * 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
US5993412A (en) * 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
US5997848A (en) * 1994-03-07 1999-12-07 Inhale Therapeutic Systems Methods and compositions for pulmonary delivery of insulin
US6005099A (en) * 1993-11-17 1999-12-21 Laboratoires Om S.A. Glucosamine disaccharides, method for their preparation, pharmaceutical composition comprising same, and their use
US6015832A (en) * 1997-12-31 2000-01-18 The Regents Of The University Of Michigan Methods of inactivating bacteria including bacterial spores
US6051256A (en) * 1994-03-07 2000-04-18 Inhale Therapeutic Systems Dispersible macromolecule compositions and methods for their preparation and use
USRE37053E1 (en) * 1996-05-24 2001-02-13 Massachusetts Institute Of Technology Particles incorporating surfactants for pulmonary drug delivery
US6294204B1 (en) * 1995-11-24 2001-09-25 Inhale Therapeutic Systems, Inc. Method of producing morphologically uniform microcapsules and microcapsules produced by this method
US20020045667A1 (en) * 1999-04-28 2002-04-18 The Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6436443B2 (en) * 1996-05-24 2002-08-20 The Penn Research Foundation, Inc. Porous particles comprising excipients for deep lung delivery
US20020119207A1 (en) * 1999-04-28 2002-08-29 The Regent Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6455673B1 (en) * 1994-06-08 2002-09-24 President And Fellows Of Harvard College Multi-mutant diphtheria toxin vaccines
US6503480B1 (en) * 1997-05-23 2003-01-07 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6506803B1 (en) * 1999-04-28 2003-01-14 Regents Of The University Of Michigan Methods of preventing and treating microbial infections
US6518239B1 (en) * 1999-10-29 2003-02-11 Inhale Therapeutic Systems, Inc. Dry powder compositions having improved dispersivity
US20030194412A1 (en) * 2001-06-05 2003-10-16 The Regents Of The University Of Michigan Nanoemulsion vaccines
US6635283B2 (en) * 1996-05-24 2003-10-21 Penn State Res Found Aerodynamically light particles for pulmonary drug delivery
US6651655B1 (en) * 2000-01-18 2003-11-25 Quadrant Technologies Limited Inhaled vaccines
US20040043041A1 (en) * 1999-04-28 2004-03-04 The Regents Of The University Of Michigan Antimicrobial compositions and methods of use
US20050238660A1 (en) * 2001-10-06 2005-10-27 Babiuk Lorne A Cpg formulations and related methods
US20050281843A1 (en) * 1999-02-26 2005-12-22 Manmohan Singh Use of bioadhesives and adjuvants for the mucosal delivery of antigens

Family Cites Families (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4436727A (en) 1982-05-26 1984-03-13 Ribi Immunochem Research, Inc. Refined detoxified endotoxin product
WO1984000111A1 (en) * 1982-06-24 1984-01-19 Robert Alan Smith Pharmaceutical gel composition
NO154957B (en) * 1984-06-27 1986-10-13 Raufoss Ammunisjonsfabrikker AIRCRAFT COVER.
CA1331443C (en) 1987-05-29 1994-08-16 Charlotte A. Kensil Saponin adjuvant
EP0468520A3 (en) 1990-07-27 1992-07-01 Mitsui Toatsu Chemicals, Inc. Immunostimulatory remedies containing palindromic dna sequences
US6165500A (en) * 1990-08-24 2000-12-26 Idea Ag Preparation for the application of agents in mini-droplets
US5389369A (en) * 1991-02-21 1995-02-14 Exoxemis, Inc. Halo peroxidase containing compositions for killing yeast and sporular microorganisms
IL101715A (en) 1991-05-02 2005-06-19 Amgen Inc Recombinant dna-derived cholera toxin subunit analogs
IT1247472B (en) 1991-05-31 1994-12-17 Fidia Spa PROCESS FOR THE PREPARATION OF MICROSPHERES CONTAINING BIOLOGICALLY ACTIVE COMPONENTS.
CA2085827C (en) 1991-12-23 2003-10-14 Lucas A. T. Hilgers Adjuvant composition containing synthetic hydrophobic lipopolysaccharide
US5968551A (en) * 1991-12-24 1999-10-19 Purdue Pharma L.P. Orally administrable opioid formulations having extended duration of effect
IT1253009B (en) 1991-12-31 1995-07-10 Sclavo Ricerca S R L DETOXIFIED IMMUNOGENIC MUTANTS OF COLERIC TOXIN AND TOXIN LT, THEIR PREPARATION AND USE FOR THE PREPARATION OF VACCINES
DK0761231T3 (en) 1992-06-25 2000-05-08 Smithkline Beecham Biolog Vaccine containing adjuvants
ES2109685T5 (en) 1993-03-23 2005-09-01 Smithkline Beecham Biologicals S.A. COMPOSITIONS FOR VACCINES CONTAINING MONOFOSFORIL-LIPIDO TO 3-O-DISABLED.
US5951988A (en) * 1993-03-30 1999-09-14 University Of Saskatchewan Adjuvant formulation with enhanced immunogenic activity, and related compositions and methods
US5576016A (en) * 1993-05-18 1996-11-19 Pharmos Corporation Solid fat nanoemulsions as drug delivery vehicles
US5961970A (en) * 1993-10-29 1999-10-05 Pharmos Corporation Submicron emulsions as vaccine adjuvants
WO1995011700A1 (en) 1993-10-29 1995-05-04 Pharmos Corp. Submicron emulsions as vaccine adjuvants
GB9326253D0 (en) 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
CN1158069C (en) 1994-05-20 2004-07-21 诺瓦瓦克斯有限公司 Antibacterial oil-in-water emulsions
US5549901A (en) * 1994-05-20 1996-08-27 Novavax, Inc. Antimicrobial oil-in-water emulsions
US5547677A (en) * 1994-05-20 1996-08-20 Novavax, Inc. Antimicrobial oil-in-water emulsions
EP1167378B1 (en) 1994-07-15 2011-05-11 University of Iowa Research Foundation Immunomodulatory oligonucleotides
AUPM873294A0 (en) 1994-10-12 1994-11-03 Csl Limited Saponin preparations and use thereof in iscoms
US5804203A (en) * 1994-12-21 1998-09-08 Cosmederm Technologies Topical product formulations containing strontium for reducing skin irritation
UA56132C2 (en) 1995-04-25 2003-05-15 Смітклайн Бічем Байолоджікалс С.А. Vaccine composition (variants), method for stabilizing qs21 providing resistance against hydrolysis (variants), method for manufacturing vaccine
WO1997029773A1 (en) 1996-02-13 1997-08-21 The Nisshin Oil Mills, Ltd. Vaccine-containing emulsion and vaccine-containing powder for oral administration and process for producing the same
US5662957A (en) * 1996-05-03 1997-09-02 Novavax, Inc. Oil containing lipid vesicles with marine applications
US5700679A (en) * 1996-06-07 1997-12-23 Novavax, Inc. Lipid vesicles having a bilayer containing a surfactant with anti-viral and spermicidal activity
US6230051B1 (en) 1996-06-18 2001-05-08 Alza Corporation Device for enhancing transdermal agent delivery or sampling
ES2241042T3 (en) 1996-10-11 2005-10-16 The Regents Of The University Of California IMMUNO STIMULATOR POLINUCLEOTIDE CONJUGATES / IMMUNOMODULATOR MOLECULA.
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
FR2755854B1 (en) * 1996-11-15 1998-12-24 Oreal NANOEMULSION BASED ON NON-IONIC AND CATIONIC AMPHIPHILIC LIPIDS AND USES
KR100453132B1 (en) 1996-12-20 2004-10-15 앨자 코포레이션 Device and method for enhancing transdermal agent flux
US6491919B2 (en) * 1997-04-01 2002-12-10 Corixa Corporation Aqueous immunologic adjuvant compostions of monophosphoryl lipid A
GB9711990D0 (en) 1997-06-11 1997-08-06 Smithkline Beecham Biolog Vaccine
ES2500490T3 (en) 1997-08-29 2014-09-30 Antigenics Inc. Compositions comprising adjuvant QS-21 and polysorbate or cyclodextrin as excipient
GB9718901D0 (en) 1997-09-05 1997-11-12 Smithkline Beecham Biolog Vaccine
AU1145699A (en) 1997-09-05 1999-03-22 Smithkline Beecham Biologicals (Sa) Oil in water emulsions containing saponins
NZ504894A (en) 1997-12-02 2002-12-20 Powderject Vaccines Inc Crystalline vaccine and adjuvant compositions jointly and separately administered to animals using a transdermal or transmucosally delivery technique
DE69810518D1 (en) * 1998-09-18 2003-02-06 Mepha Ag Aesch TOPICALLY ADMINISTRABLE ALKYL, PHENYL PYRIDONE MEDICINAL PRODUCTS
WO2000037105A2 (en) 1998-12-21 2000-06-29 Medimmune, Inc. Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
CZ302790B6 (en) 1998-12-23 2011-11-09 Id Biomedical Corporation Isolated polynucleotide, vector, host cell, production process, isolated polypeptide, chimeric polypeptide, vaccine and use thereof
ES2242604T3 (en) 1999-02-26 2005-11-16 Chiron Corporation MICROEMULSIONS WITH MACROPARTICLES AND ADSORBID MICROPARTICLES.
US8236335B2 (en) * 1999-04-28 2012-08-07 The Regents Of The University Of Michigan Antimicrobial nanoemulsion compositions and methods
US7655252B2 (en) * 1999-04-28 2010-02-02 The Regents Of The University Of Michigan Antimicrobial nanoemulsion compositions and methods
US6558695B2 (en) * 1999-12-16 2003-05-06 Dermatrends, Inc. Topical and transdermal administration of peptidyl drugs using hydroxide releasing agents as permeation enhancers
US20020155084A1 (en) * 2000-06-02 2002-10-24 The Regents Of The University Of The Michigan Nanoemulsion formulations
IL153558A0 (en) 2000-06-20 2003-07-06 Shire Biochem Inc Streptococcus antigens
US6565873B1 (en) * 2000-10-25 2003-05-20 Salvona Llc Biodegradable bioadhesive controlled release system of nano-particles for oral care products
US20060134186A1 (en) * 2002-03-28 2006-06-22 Carlton Richard M Oxygenating agents for enhancing host responses to microbial infections
US8946295B2 (en) * 2002-07-25 2015-02-03 Sunny Pharmtech Inc. Histone hyperacetylating agents for promoting wound healing and preventing scar formation
US20060233721A1 (en) * 2002-10-25 2006-10-19 Foamix Ltd. Foam containing unique oil globules
US9211259B2 (en) * 2002-11-29 2015-12-15 Foamix Pharmaceuticals Ltd. Antibiotic kit and composition and uses thereof
US20070292355A1 (en) * 2002-10-25 2007-12-20 Foamix Ltd. Anti-infection augmentation foamable compositions and kit and uses thereof
US20080138296A1 (en) * 2002-10-25 2008-06-12 Foamix Ltd. Foam prepared from nanoemulsions and uses
US9265725B2 (en) * 2002-10-25 2016-02-23 Foamix Pharmaceuticals Ltd. Dicarboxylic acid foamable vehicle and pharmaceutical compositions thereof
US20050208083A1 (en) * 2003-06-04 2005-09-22 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositons, and methods of use thereof
CA2528095C (en) * 2003-06-04 2014-10-14 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositions, and methods of use thereof
PL1648412T3 (en) * 2003-07-07 2008-04-30 Nares Ab Microemulsions and its use for preventing airway diseases
JP3808846B2 (en) * 2003-07-07 2006-08-16 独立行政法人海上技術安全研究所 Fatigue crack growth suppression method and detection method, and paste used therefor
WO2005030172A1 (en) * 2003-09-24 2005-04-07 The Regents Of The University And Methods Antimicrobial nanoemulsion compositions and methods
US9028852B2 (en) * 2004-09-07 2015-05-12 3M Innovative Properties Company Cationic antiseptic compositions and methods of use
US20060058238A1 (en) * 2004-09-15 2006-03-16 Lee Laurent-Applegate Fetal skin cell protein compositions for the treatment of skin conditions, disorders or diseases and methods of making and using the same
US8197851B2 (en) * 2004-09-29 2012-06-12 Michael Ary Bos Carotenoid composition and method for preparation thereof
DE102004049574A1 (en) * 2004-10-12 2006-04-20 Doris Dr. Barnikol-Keuten Drugs and System for Percutaneous Drug Delivery
US20060204469A1 (en) * 2005-03-09 2006-09-14 Eric Spengler Stable mixed emulsions
EP1871349A1 (en) * 2005-04-11 2008-01-02 Nanobio Corporation Quaternary ammonium halides for treatment of infectious conditions
CA2618974C (en) * 2005-08-09 2014-01-28 Nanobio Corporation Nanoemulsion compositions having anti-inflammatory activity
US20070292688A1 (en) * 2005-08-18 2007-12-20 Eastman Kodak Company Silylamine modified nanoparticulate carriers
US20070105754A1 (en) * 2005-11-10 2007-05-10 Pinto Da Silva Luis L Pharmaceutical composition to prevent and treat epithelial wounds, immunomodulating pharmaceutical composition, pharmaceutical composition, insecticide, insecticide composition insecticide use of lectin KM+ to treat cicatrizations, use of lectin KM+ to prepare immunomodulating medicament, use of lectin KM+ to prepare anti-bacterial medicament, use lectin KM+ to prpare anti-viral medicament, use of lectin KM+ to prepare anti-parasite medicament, use of lectin KM+ to prepare anti-fungal medicament, use of lectin KM+ to prepare anti-parasite medicament expression method, DNA vector, recombinant
WO2007106555A2 (en) * 2006-03-14 2007-09-20 Neuren Pharmaceuticals Limited Oral formulations of glycyl-2-methylprolyl-glutamate
CA2653390C (en) * 2006-06-02 2014-07-08 Hawaii Chitopure, Inc. Chitosan-derivative compounds and methods of controlling microbial populations
DE102006047529A1 (en) * 2006-10-07 2008-04-10 Evonik Goldschmidt Gmbh Dermatological composition contains an oligopeptide as an agent for increasing skin sensitivity
US20080206155A1 (en) * 2006-11-14 2008-08-28 Foamix Ltd. Stable non-alcoholic foamable pharmaceutical emulsion compositions with an unctuous emollient and their uses
GB0701021D0 (en) * 2007-01-19 2007-02-28 Aquapharm Bio Discovery Ltd Inducion of microbial secondary metabolites
DE102007007698A1 (en) * 2007-02-09 2008-08-21 Exner, Heinrich, Dr. Use of a highly emulsified silicone oil emulsion (HSE) for the manufacture of a medicament
US8747872B2 (en) * 2007-05-02 2014-06-10 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
US9161943B2 (en) * 2007-12-31 2015-10-20 Industrial Technology Research Institute Sustained release composition and manufacturing method thereof
RU2547441C2 (en) * 2008-03-06 2015-04-10 Анакор Фармасьютикалз, Инк. Boron-containing small molecules as anti-inflammatory agents
ES2628407T3 (en) * 2008-04-01 2017-08-02 Antipodean Pharmaceuticals, Inc. Compositions and procedures for skin care
JP2011521896A (en) * 2008-04-15 2011-07-28 サーコード コーポレイション Topical LFA-1 antagonists for use in local treatment for immune related disorders
CA2721800A1 (en) * 2008-04-21 2009-10-29 Nanobio Corporation Nanoemulsion influenza vaccine
CA2722445A1 (en) * 2008-04-25 2009-10-29 Nanobio Corporation Nanoemulsions for treating fungal, yeast and mold infections
AU2009293595A1 (en) * 2008-05-23 2010-03-25 The Regents Of The University Of Michigan Nanoemulsion adjuvants
US9415006B2 (en) * 2008-05-23 2016-08-16 The Regents Of The University Of Michigan Immunogenic compositions comprising nanoemulsion and hepatitis B virus immunogen and methods of using the same
US20100068162A1 (en) * 2008-09-18 2010-03-18 Lipo Chemicals Inc. Compositions Having Silicone Properties
US20100092526A1 (en) * 2008-09-26 2010-04-15 Nanobio Corporation Nanoemulsion therapeutic compositions and methods of using the same
EP2393491B1 (en) * 2009-02-03 2020-04-22 Microbion Corporation Bismuth-thiols as antiseptics for epithelial tissues, acute and chronic wounds, bacterial biofilms and other indications
US8464435B2 (en) * 2011-02-25 2013-06-18 Deere & Company Offset check gauge for flexible bellows

Patent Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3854480A (en) * 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US4675189A (en) * 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
US4452775A (en) * 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
US4596556A (en) * 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4709017A (en) * 1985-06-07 1987-11-24 President And Fellows Of Harvard College Modified toxic vaccines
US4950740A (en) * 1987-03-17 1990-08-21 Cetus Corporation Recombinant diphtheria vaccines
US5057540A (en) * 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US4895452A (en) * 1988-03-03 1990-01-23 Micro-Pak, Inc. Method and apparatus for producing lipid vesicles
US5133974A (en) * 1989-05-05 1992-07-28 Kv Pharmaceutical Company Extended release pharmaceutical formulations
US5103497A (en) * 1989-11-14 1992-04-07 Hicks John W Flying spot endoscope
US5284656A (en) * 1991-03-15 1994-02-08 Amgen Inc. Pulmonary administration of granulocyte colony stimulating factor
US5565332A (en) * 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5407686A (en) * 1991-11-27 1995-04-18 Sidmak Laboratories, Inc. Sustained release composition for oral administration of active ingredient
US5843711A (en) * 1992-05-06 1998-12-01 The Regents Of The University Of California Diphtheria toxin receptor-binding region
US5592904A (en) * 1993-05-03 1997-01-14 Negre; Guy Method and devices for controlling the combustion of a four stroke engine
US6005099A (en) * 1993-11-17 1999-12-21 Laboratoires Om S.A. Glucosamine disaccharides, method for their preparation, pharmaceutical composition comprising same, and their use
US5997848A (en) * 1994-03-07 1999-12-07 Inhale Therapeutic Systems Methods and compositions for pulmonary delivery of insulin
US6423344B1 (en) * 1994-03-07 2002-07-23 Inhale Therapeutic Systems Dispersible macromolecule compositions and methods for their preparation and use
US6051256A (en) * 1994-03-07 2000-04-18 Inhale Therapeutic Systems Dispersible macromolecule compositions and methods for their preparation and use
US5451569A (en) * 1994-04-19 1995-09-19 Hong Kong University Of Science And Technology R & D Corporation Limited Pulmonary drug delivery system
US5917017A (en) * 1994-06-08 1999-06-29 President And Fellows Of Harvard College Diphtheria toxin vaccines bearing a mutated R domain
US6455673B1 (en) * 1994-06-08 2002-09-24 President And Fellows Of Harvard College Multi-mutant diphtheria toxin vaccines
US5736152A (en) * 1995-10-27 1998-04-07 Atrix Laboratories, Inc. Non-polymeric sustained release delivery system
US6294204B1 (en) * 1995-11-24 2001-09-25 Inhale Therapeutic Systems, Inc. Method of producing morphologically uniform microcapsules and microcapsules produced by this method
USRE37053E1 (en) * 1996-05-24 2001-02-13 Massachusetts Institute Of Technology Particles incorporating surfactants for pulmonary drug delivery
US5985309A (en) * 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
US6436443B2 (en) * 1996-05-24 2002-08-20 The Penn Research Foundation, Inc. Porous particles comprising excipients for deep lung delivery
US6635283B2 (en) * 1996-05-24 2003-10-21 Penn State Res Found Aerodynamically light particles for pulmonary drug delivery
US6447753B2 (en) * 1996-05-24 2002-09-10 The Penn Research Foundation, Inc. Porous particles for deep lung delivery
US5993412A (en) * 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
US6503480B1 (en) * 1997-05-23 2003-01-07 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6015832A (en) * 1997-12-31 2000-01-18 The Regents Of The University Of Michigan Methods of inactivating bacteria including bacterial spores
US20050281843A1 (en) * 1999-02-26 2005-12-22 Manmohan Singh Use of bioadhesives and adjuvants for the mucosal delivery of antigens
US20020119207A1 (en) * 1999-04-28 2002-08-29 The Regent Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6559189B2 (en) * 1999-04-28 2003-05-06 Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6506803B1 (en) * 1999-04-28 2003-01-14 Regents Of The University Of Michigan Methods of preventing and treating microbial infections
US6635676B2 (en) * 1999-04-28 2003-10-21 Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US20040043041A1 (en) * 1999-04-28 2004-03-04 The Regents Of The University Of Michigan Antimicrobial compositions and methods of use
US20020045667A1 (en) * 1999-04-28 2002-04-18 The Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6518239B1 (en) * 1999-10-29 2003-02-11 Inhale Therapeutic Systems, Inc. Dry powder compositions having improved dispersivity
US6651655B1 (en) * 2000-01-18 2003-11-25 Quadrant Technologies Limited Inhaled vaccines
US20030194412A1 (en) * 2001-06-05 2003-10-16 The Regents Of The University Of Michigan Nanoemulsion vaccines
US20050238660A1 (en) * 2001-10-06 2005-10-27 Babiuk Lorne A Cpg formulations and related methods

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Caraher, E, G Reynolds, P Murphy, S McClean, and M Callaghan. 2006. Comparison of antibiotic susceptibility of Burkholderia cepacia complex organisms when grown planktonically or as biofilm in vitro. Eur. J. Clin. Microbiol. Infect. Dis; 26: 213-216. *

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100278812A1 (en) * 2001-06-22 2010-11-04 Cpex Pharmaceuticals, Inc. Nasal immunization
US8481043B2 (en) 2001-06-22 2013-07-09 Cpex Pharmaceuticals, Inc. Nasal immunization
US20100203139A1 (en) * 2008-09-26 2010-08-12 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
US9259407B2 (en) 2008-09-26 2016-02-16 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
US8962026B2 (en) 2008-09-26 2015-02-24 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
US20140193461A1 (en) * 2009-05-14 2014-07-10 The Regents Of The University Of Michigan Streptococcus vaccine compositions and methods of using the same
WO2012003361A3 (en) * 2010-07-02 2012-04-12 The Regents Of The University Of Michigan Nanoemulsion vaccines
JP2016168058A (en) * 2010-10-28 2016-09-23 栄研化学株式会社 Disk test specimen for detecting drug-resistant bacterium
WO2012078482A1 (en) * 2010-12-10 2012-06-14 Merck Sharp & Dohme Corp. Novel formulations which mitigate agitation-induced aggregation of immunogenic compositions
CN103391714A (en) * 2010-12-10 2013-11-13 默沙东公司 Novel formulations which mitigate agitation-induced aggregation of immunogenic compositions
US9139616B2 (en) 2010-12-20 2015-09-22 University Of Utah Research Foundation Isolation, identification, and uses of antifungal compounds
US9879048B2 (en) 2010-12-20 2018-01-30 University Of Utah Research Foundation Isolation, identification, and uses of antifungal compounds
US8785376B2 (en) 2010-12-20 2014-07-22 Virginia Tech Intellectual Properties Isolation, identification, and uses of antifungal compounds
US20170347661A1 (en) * 2011-04-06 2017-12-07 Convatec Technologies Inc. Composition comprising antimicrobial metal ions and a quaternary cationic surfactant
US9572774B2 (en) 2011-05-19 2017-02-21 Savara Inc. Dry powder vancomycin compositions and associated methods
US10561608B2 (en) 2011-05-19 2020-02-18 Savara Inc. Dry powder Vancomycin compositions and associated methods
EP3329936A1 (en) 2011-08-22 2018-06-06 Nanobio Corporation Herpes simplex virus nanoemulsion vaccine
US11147869B2 (en) 2011-08-22 2021-10-19 Bluewillow Biologics, Inc. Herpes simplex virus nanoemulsion vaccine
US10206996B2 (en) 2011-08-22 2019-02-19 Nanobio Corporation Herpes simplex virus nanoemulsion vaccine
US9561271B2 (en) 2011-09-09 2017-02-07 Nanobio Corporation Nanoemulsion respiratory syncytial virus (RSV) subunit vaccine
US10596251B2 (en) 2011-09-09 2020-03-24 Nanobio Corporation Nanoemulsion respiratory syncytial virus (RSV) subunit vaccine
WO2013096697A3 (en) * 2011-12-20 2015-06-18 University Of Utah Research Foundation Isolation, identification, and used of antifungal compounds
US20150216902A1 (en) * 2012-07-27 2015-08-06 Kassem Khal Fardoussi PHARMACEUTICAL COMPOSITION OF MILK FAT GLOBULES (MFGs) ZINC FREE
WO2014151449A1 (en) * 2013-03-15 2014-09-25 Purdue Research Foundation Antimicrobial compositions and methods of use
CN105705014A (en) * 2013-03-15 2016-06-22 普渡研究基金会 Antimicrobial compositions and methods of use
US10383330B2 (en) 2013-03-15 2019-08-20 Purdue Research Foundation Antimicrobial compositions and methods of use
US20180008711A1 (en) * 2015-02-05 2018-01-11 Marc Selner Triple antibiotic cationic ointment
US20160368953A1 (en) * 2015-06-18 2016-12-22 The United States of America, as represented by the Secretary of Agirculture Bacteriocin with novel activity
US9598471B2 (en) * 2015-06-18 2017-03-21 The United States Of America, As Represented By The Secretary Of Agriculture Bacteriocin with novel activity
US10722461B2 (en) 2015-06-19 2020-07-28 Global Health Solutions, Llc Petrolatum-based delivery systems and for active ingredients
US10675243B2 (en) 2015-06-19 2020-06-09 Global Health Solutions Llc Petrolatum-based compositions comprising cationic biocides
US10874608B2 (en) 2015-06-19 2020-12-29 Global Health Solutions Llc Petrolatum-based compositions comprising cationic biocides
WO2017017625A1 (en) * 2015-07-27 2017-02-02 Sun Pharmaceutical Industries Limited A stable topical pharmaceutical composition comprising nanonized silver sulfadiazine
US10729698B2 (en) * 2015-11-03 2020-08-04 Zoetis Services Llc Sol-gel polymer composites and uses thereof
US11654112B2 (en) 2016-11-22 2023-05-23 Elektrofi, Inc. Particles comprising a therapeutic or diagnostic agent and suspensions and methods of use thereof
WO2018098376A1 (en) * 2016-11-22 2018-05-31 Elektrofi, Inc. Particles comprising a therapeutic or diagnostic agent and suspensions and methods of use thereof
US11252960B2 (en) 2017-01-31 2022-02-22 Kimberly-Clark Worldwide, Inc. Antibacterial composition including benzoic acid ester and methods of inhibiting bacterial growth utilizing the same
US11795192B2 (en) 2017-03-10 2023-10-24 Lakewood Amedex, Inc. Antimicrobial compounds, compositions, and uses thereof
WO2019055370A1 (en) * 2017-09-12 2019-03-21 Academic Pharmaceuticals, Inc. Metolazone emulsion formulation
US11389451B2 (en) * 2017-09-12 2022-07-19 Hyloris Developments S.A. Metolazone emulsion formulation
US20210330645A1 (en) * 2018-09-07 2021-10-28 Massey University Antibacterial combinations
US11318149B2 (en) * 2018-09-27 2022-05-03 Board Of Trustees Of Michigan State University Compositions and methods for inhibiting biofilm-forming bacteria
CN110333216A (en) * 2018-12-29 2019-10-15 厦门市普识纳米科技有限公司 The detection method of xacin-series antibiotic in a kind of animal foodstuff
US11717488B2 (en) 2019-01-31 2023-08-08 Elektrofi, Inc. Particle formation and morphology
WO2020240472A1 (en) * 2019-05-28 2020-12-03 Aybar Ecotechnologies Corp. Wide-spectrum antibacterial pharmaceutical formulations comprising lysozyme and methods of using the same
US11459376B2 (en) 2019-09-13 2022-10-04 Elektrofi, Inc. Compositions and methods for the delivery of therapeutic biologics for treatment of disease
CN112569187A (en) * 2019-09-27 2021-03-30 武汉科福新药有限责任公司 Tacrolimus nano eye emulsion and preparation method thereof
NL2027383A (en) 2020-01-24 2021-09-01 Aim Immunotech Inc Methods, compositions, and vaccines for treating a virus infection
WO2021262728A1 (en) * 2020-06-23 2021-12-30 Virogenomics Biodevelopment, Inc. Compositions and methods for viral detection
CN114246839A (en) * 2020-09-22 2022-03-29 鲁南制药集团股份有限公司 Oral nanoemulsion enteric capsule and preparation method thereof
CN115007119A (en) * 2021-03-03 2022-09-06 中国科学院大连化学物理研究所 Zwitterion-containing hydrophilic microsphere and preparation and application thereof

Also Published As

Publication number Publication date
US20100203139A1 (en) 2010-08-12
US8962026B2 (en) 2015-02-24
US20150216826A1 (en) 2015-08-06
WO2010036938A2 (en) 2010-04-01
US9259407B2 (en) 2016-02-16
JP5722782B2 (en) 2015-05-27
AU2009296458A1 (en) 2010-04-01
WO2010036938A3 (en) 2010-06-10
CA2738245A1 (en) 2010-04-01
US20160158179A1 (en) 2016-06-09
WO2010036945A2 (en) 2010-04-01
EP2349209A2 (en) 2011-08-03
CN102223876A (en) 2011-10-19
WO2010036945A3 (en) 2010-05-20
JP2012504134A (en) 2012-02-16

Similar Documents

Publication Publication Date Title
US9259407B2 (en) Nanoemulsion therapeutic compositions and methods of using the same
US8877208B2 (en) Multivalent nanoemulsion vaccines
US11806318B2 (en) Nanoemulsion compositions for preventing, suppressing or eliminating allergic and inflammatory disease
US20090291095A1 (en) Nanoemulsion adjuvants
US20190021998A1 (en) Topical nanoemulsion therapy for wounds
JP6122083B2 (en) Nanoemulsion vaccine
US20140093537A1 (en) Immunogenic compositions comprising nanoemulsion and methods of administering the same
US20130273113A1 (en) Immunogenic apoptosis inducing compositions and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: NANOBIO CORPORATION,MICHIGAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BAKER, JR., JAMES R.;HAMOUDA, TAREK;SUTCLIFFE, JOYCE A.;SIGNING DATES FROM 20091123 TO 20091130;REEL/FRAME:023745/0197

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION