CA2210996C - Liquid formulations for proteinic pharmaceuticals - Google Patents

Liquid formulations for proteinic pharmaceuticals Download PDF

Info

Publication number
CA2210996C
CA2210996C CA002210996A CA2210996A CA2210996C CA 2210996 C CA2210996 C CA 2210996C CA 002210996 A CA002210996 A CA 002210996A CA 2210996 A CA2210996 A CA 2210996A CA 2210996 C CA2210996 C CA 2210996C
Authority
CA
Canada
Prior art keywords
sodium
pharmaceutical agent
combination
polyoxyethylene
deoxycholate
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.)
Expired - Fee Related
Application number
CA002210996A
Other languages
French (fr)
Other versions
CA2210996A1 (en
Inventor
Pankaj Modi
Subash Chandarana
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2210996A1 publication Critical patent/CA2210996A1/en
Application granted granted Critical
Publication of CA2210996C publication Critical patent/CA2210996C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/28Steroids, e.g. cholesterol, bile acids or glycyrrhetinic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

A liquid pharmaceutical agent formulation suitable for oral or nasal delivery comprises a proteinic pharmaceutical agent, water and at least two absorption enhancing compounds. The absorption enhancing compounds are selected from sodium salicylate, sodium lauryl sulphate, disodium ethylenediaminetetraacetic acid (disodium EDTA), oleic acid, linoleic acid, monoolein, lecithin, lysolecithin, deoxycholate, sodium deoxycholate, chenodeoxycholate, taurodeoxycholate, glycochenodeoxycholate, polyoxyethylene X-lauryl ether wherein X is from 9 to 20, sodium tauro-24, 25-dihydrofusidate, polyoxyethylene ether, polyoxyethylene sorbitan esters, p-t-octylphenoxypolyoxyethylene, N-lauryl-.beta.-D-maltopyranoside, 1-dodecylazacycloheptane-2-azone and phospholipids, wherein the amount of each of the absorption enhancing compounds is present in a concentration of from 1 to 10 wt./wt.% of the total formulation. Preferably each of the absorption enhancing compounds is present in a concentration of from 1.5 to 3.5 wt./wt.% The formulation is particulary adapted to oral delivery of insulin. A preferred insulin formulation contains about 2 wt.% each of chenodeoxycholate, deoxycholate and polyoxyethylene 9-lauryl ether absorption enhancers, an inorganic salt, e.g. sodium chloride, a protective polymer, e.g. geletin, a protease inhibitor, e.g. bacitracin, and optionally an antioxidant, e.g. tocopherol.

Description

.
Liquid Formulations for Proteinic Pharmaceuticals Technical Field The present invention relates to an improved delivery system for the administration of pharmaceuticals, e.g. peptidic drugs, vaccines and hormones. In particular it relates to insulin, vaccines and hormones which may be orally administered.
Backqround In the treatment of diabetes, current subcutaneous insulin therapy is limited by the delayed time of onset and time of peak action for the insulin, inability to accommodate changing insulin requirements, e.g. during exercise or meals, and by the large inter- and intra-subject coefficient of variation of absorption and insulin action. Oral delivery of insulin would overcome many of the disadvantages of the subcutaneous delivery system.
The inability of subcutaneous insulin delivery to effectively and safely control glucose levels has encouraged exploration of alternate, less painful methods of delivery that might provide a faster rate of insulin absorption and a relatively short half life.
Oral administration of insulin to treat diabetes is likely to be attractive because of its virtual lack of toxicity and its inherent clinical applicability in the reduction in severity of lymphocytic infiltration of pancreatic islets. Furthermore, it has been shown that splenic T-cells from animals orally treated with insulin adoptively transfer protection against diabetes, which indicates that oral insulin generates active cellular mechanisms that suppress disease. Such results suggest that oral insulin affects diabetes and the pancreatic cellular inflammatory process and raises the possibility that oral administration of insulin may provide a new approach for the treatment of autoimmune diabetes.
As indicated, oral delivery of insulin would have many benefits. Oral delivery is also a preferred method of delivering many other therapeutic or pharmaceutical agents. In some instance, even nasal delivery is preferred from a patient point of view because it is not painful like subcutaneous administration and it may be easily self-administered. Nasal delivery of therapeutic agents has the disadvantage that the amount of agent (dose) delivered varies from one dose to another, for a variety of reasons. For example, the lining of the nose is sensitive and sneezing or dripping as a result of irritation of the lining causes loss of usable therapeutic agent. Oral delivery of therapeutic or pharmaceutical agents overcome these difficulties. Even so, if a nasal delivery method is desired, an improved formulation would be helpful.
Effective oral delivery of a pharmaceutical agent requires that the agent has sufficient solubility in the stomach and intestinal lumen and the ability to pass through the intestinal wall. Many peptidic drugs have extremely poor absorption in the gastrointestinal tract and tend to degrade quickly. For example insulin, when introduced orally, has extremely poor absorption in the gastrointestinal tract, tends to degrade quickly, and thus has no metabolic effect on blood glucose levels.
M. Kidron, H. Bar-on, E.M. serry and E. Ziv in Life Sciences, vol 29, pp 803-9 (1981) and vol 31, pp 2837-41 (1982) have experimented in small animals, with surgical delivery of insulin to the small intestine of a composition of 5 wt.% of an absorption enhancer, e.g.
sodium cholate, 2 wt% soyabean trypsin inhibitor and 15 IU of insulin. Such a composition had a metabolic effect on blood sugar level, i.e reduced the blood sugar level by about 30%. However a large amount of insulin was required to produce this effect. The method is obviously not practical in humans and the amount of insulin required would be prohibitively expensive.
A composition which provides effective and practical oral or for some compositions, nasal delivery of pharmaceutical agents has been found.
Disclosure of Invention Accordingly the present invention provides a liquid pharmaceutical agent formulation suitable for oral or nasal delivery comprising a proteinic pharmaceutical agent, water and at least two absorption enhancing compounds selected from the group consisting of sodium salicylate, sodium lauryl sulphate, disodium ethylenediaminetetraacetic acid (disodium EDTA), oleic acid, linoleic acid, monoolein, lecithin, lysolecithin, deoxycholate, sodium deoxycholate, chenodeoxycholate, taurodeoxycholate, glycochenodeoxycholate, polyoxyethylene X-lauryl ether wherein X is from 9 to 20, sodium tauro-24, 25-dihydrofusidate, polyoxyethylene ether, polyoxyethylene sorbitan esters, p-t-octylphenoxypolyoxyethylene, N-lauryl-~-D-maltopyranoside, 1-dodecylazacycloheptane-2-azone and phospholipids, wherein the amount of each of the absorption enhancing compounds is present in a concentration o-f from 1 to 10 wt./wt.% of the total formulation.
Sodium lauryl sulphate is sometimes referred to as sodium dodecylsulphate.
In a preferred embodiment the concentration of each of the absorption enhancing compounds is from 1 to 5 wt./wt.% and especially from 1.5 wt./wt.% to 3.5 wt./wt.%
In a preferred embodiment the absorption enhancing compounds are selected from a combination of deoxycholate, chenodeoxycholate, and polyoxyethylene 9-lauryl ether, a combination of sodium salicylate, deoxycholate, chenodeoxycholate, and polyoxyethylene 9-lauryl ether, a combination of sodium deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and monoolein, a combination of deoxycholate, chenodeoxycholate and sodium salicylate, a combination of deoxycholate, sodium salicylate and sodium lauryl sulphate, a combination of deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and sodium tauro-24, 25-dihydrofusidate, a combination of sodium deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and sodium tauro-24, 25-dihydrofusidate, a combination of deoxycholate, chenodeoxycholate, taurodeoxycholate, polyoxyethylene 9-lauryl ether and monoolein, a combination of chenodeoxycholate, glycochenodeoxycholate, polyoxyethylene 9-lauryl ether and sodium tauro-24, 25-dihydrofusidate, a combination of chenodeoxycholate, sodium lauryl sulphate and disodium EDTA, a combination of deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and disodium EDTA, a combination of sodium salicylate, disodium EDTA and polyoxyethylene 9-lauryl ether, a combination of monoolein, oleic acid and polyoxyethylene sorbitan ester, a combination of monoolein, oleic acid, polyoxyethylene sorbitan ester and sodium lauryl sulphate, and a combination of linoleic acid, monoolein and sodium salicylate.
A preferred polyoxyethylene sorbitan ester is available under the trade mark Tween 80.
In one embodiment one of the enhancing compounds is polyoxyethylene X-lauryl ether, wherein X is 9 or 10.
In another embodiment the phospholipid is selected from the group consisting of lecithin, lysolecithin, sphingomyelin, phosphatidylcholine, cephalin and phosphatidylethanolamine.
The most preferred absorption enhancing compounds are deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether, sodium salicylate, monoolein and sodium tauro-24, 25-dihydrofusidate. These in particular, when combined with each other, or with others of the named absorption enhancing compounds, are the most effective.
Preferred combinations are i) deoxycholate, chenodeoxycholate and sodium salicylate, ii) sodium salicylate, deoxycholate and sodium lauryl sulphate, .

iii) chenodeoxycholate, sodium lauryl sulphate and disodium EDTA and iv) monoolein, oleic acid and polyoxyethylene sorbitan ester, v) deoxycholate, chenodeoxycholate and polyoxyethylene 9-lauryl ether.
The most preferred combination is deoxycholate, chenodeoxycholate and polyoxyethylene 9-lauryl ether.
For insulin-containing compositions, it is preferred that the composition also contains at least one inorganic salt which opens channels in the gastrointestinal tract and may provide additional stimulation to release insulin. Non-limiting examples of inorganic salts are sodium, potassium, calcium and zinc salts, especially sodium chloride, potassium chloride, calcium chloride, zinc chloride and sodium bicarbonate.
It is also preferred that the composition contain at least one protective polymer for slow release of the pharmaceutical agent. Preferred protective polymers are polyvinyl alcohol, polyethylene glycol, and gelatin.
It will be recognized by those skilled in the art that for many pharmaceutical compositions it is usual to add at least one antioxidant to prevent degradation and oxidation of the pharmaceutically active ingredients.
It will also be understood by those skilled in the art that colorants, flavouring agents and non-therapeutic amounts of other compounds may be included in the formulation.
In one embodiment the antioxidant is selected from the group consisting of tocopherol, deteroxime mesylate, methyl paraben and ascorbic acid.
In yet another embodiment the antioxidant is present in an amount of 1 to 3 g for every litre of water in the formulation. A preferred antioxidant is tocopherol.
In a preferred embodiment at least one protease inhibitor is added to the formulation to inhibit degradation of the pharmaceutical agent by the action of proteolytic enzymes. Of the known protease inhibitors, most are effective at concentrations of from 1 to 3 wt./wt.% of the formulation.
Non-limiting examples of effective protease inhibitors are bacitracin, soyabean trypsin and aprotinin. Bacitracin is the most effective of the three named herein when used in concentrations of from 1.5 to 2.0 wt./wt.%. The other two may be used in concentrations of about 1 to 2 wt./wt.%.
The pharmaceutical agents may be selected from insulin, antigens selected from the group consisting of MMR (mumps, measles and rubella~ vaccine, typhoid vaccine, hepatitis A vaccine, hepatitis B vaccine, herpes simplex virus, bacterial toxoids, cholera toxin B-subunit, influenza vaccine virus, whooping cough vaccine virus, vaccinia virus, adenovirus, canary pox, polio vaccine virus, malaria vaccine virus, bacillus calmette geurin (BCG), klebsiella pneumoniae, HIV
envelop glycoproteins and cytokins and other agents selected from the group consisting of bovine somatropine (sometimes referred to as sST), estrogens, androgens, insulin growth factors (sometimes referred to as IGF), interleukin-I, interleukin-II and cytokins. Three such cytokins are interferon-~, interferon-y and tuftsin.
Examples of bacterial toxoids are tetanus, diphtheria, pseudomonas A, mycobacterium tuberculosis.
Examples of HIV envelop glycoproteins are gp 120 and gp 160 for AIDS vaccines. Examples of anti-ulcer H~
receptor antagonists are ranitidine, cimetidine and famotidine, and other anti-ulcer drugs are omparazide, cesupride and misoprostol. An example of a hypoglycaemic agent is glizipide. Insulin is used for the control of diabetes.
As will be understood by those skilled in the art, two or more pharmaceutical agents may be combined for specific effects. The necessary amounts of active ingredient can be determined by simple experimentation.

, Specific pharmaceutical agents which are particularly suited to this invention are insulin, heparin, low molecular weight heparin, hirugen, hirulos and huridine.
In a further embodiment the pharmaceutical agent is insulin.
The method of making the formulation is easy.
Typically the absorption enhancing compounds are added to cold water and vigorously mixed. The pharmaceutical agent or agents, any antioxidants, inorganic salts, protective polymers, protease inhibitors and other ingredients, e.g. colouring matter, flavourings, are then added and mixed until the solution is homogeneous.
In the selection of a suitable absorption enhancing compound combination, it has been found that the amount of total absorption enhancing compound should be less than about 10 wt./wt.% of the formulation and preferably from 1 to 5 wt./wt.%. Frequent use or prolonged use of higher concentrations of absorption enhancing compounds are likely to be harmful to linings and tissues in the gastrointestinal tract, and may cause diarrhoea. It is believed that the optimum range for most combinations is from 1.5 to 3.5 wt./wt.%
In general, advantages of the present formulation are that adverse reactions are decreased and bioavailability is increased. Furthermore, manufacture of the formulation is simple. Du~ to the liquid nature of the formulation, ingestion is easy, the action of the pharmaceutical agent can occur very rapidly, e.g. within 10-15 minutes, and the problem of gastric emptying is minimized.
The invention is now exemplified by reference to insulin, although this is not to be taken as limiting.
In the case of insulin, oral administration provides for rapid onset of action, gives a long duration of action, is convenient and is free from the discomfiture of injection. In addition, it more closely mimics normal physiological insulin secretion. The present oral insulin may serve as the sole substitution of insulin injection in the case of insulin dependent diabetes mellitus and pharmacologic therapy for non-insulin dependent diabetes mellitus.
The invention is illustrated by the following non-limiting examples.
Example I
0.2 g of chenodeoxycholate, 0.2 g of deoxycholate, 0.2 g polyoxyethylene 9-lauryl ether were dissolved in a small quantity of cold (4~C) distilled water which contained 0.2% gelatin and 0.9~ sodium chloride.
Dissolution was effected with rapid stirring, and the volume topped up to 9 ml with distilled water. To this clear suspension was added 1 ml of Novolin-R (trade mark) fast acting insulin. The mixture was stirred continuously to give a homogenous solution. One millilitre aliquots of this solution (10 IU insulin) were then administered orally to diabetic rats according to the following protocol. Several batches were made in order to feed to the number of rats in the study.
Fifty-five genetically diabetic (urine glucose positive) male wistar BB strain rats weighing 300-400 g were used in the study. The rats were grouped in five groups of eleven rats each. The rats were monitored for diabetes by blood glucose testing using an Accucheck-III
(trade mark) glucometer. Diabetes was confirmed by severe hyperglycaemia, with blood glucose levels of greater than 30 mmol/L.
At the beginning of the experiment, each rat was numbered and bled for a blood glucose level measurement.
Each rat was gavaged with a 1 ml solution containing 3 parts Hanks balanced salt solution to 7 parts sodium bicarbonate to neutralize the stomach acid. One millilitre portions of the oral insulin formulation prepared as above was mixed with 1 ml of saline solution portions and then administered to the rats with a stainless steel gavage tube. One dose was administered per rat per day. Blood was removed via an implanted angiocatheter in a tail vein for determination of serum insulin levels at 0, 2, 5, 10, lS 30, 45, 60, 90 and 120 minutes after administration of the dose. The resultant sera were frozen and sent to a laboratory for analyzing total serum insulin levels by radioimmunoassay. In addition, 10 yl blood samples were drawn from each rat at 0, 30, 60, 120, 180, 240, 300, 360 minutes and 24 hours after administration of the dose, for glucose level analysis. The results for 22 of the rats are shown in Table I, and for 5 of the rats are shown in Table II.
Table I, Changes in blood glucose levels (mmol/L):
Rat Time (hours' 0 0.5 1.0 2.0 3.0 5.0 6.0 24.0 1 30.0 21.2 13.110.3 12.4 16.5 18.2 19.7 2 29.5 26.0 12.811.0 13.1 17.4 16.9 18.1 3 28.7 25.5 21.5 7.7 12.8 16.1 21.8 18.5 4 28.0 19.1 13.410.1 15.3 19.8 20.8 20.0 30.8 18.0 12.110.0 12.3 13.0 21.7 22.2 6 28.6 13.2 10.1 9.2 13.4 15.3 18.8 16.1 7 28.9 23.4 18.0 6.2 11.8 16.6 22.6 20.6 8 30.1 18.3 10.4 7.2 9.6 17.1 23.2 21.3 9 26.8 12.1 10.7 6.9 13.8 16.8 18.7 17.0 27.3 18.1 11.5 6.4 11.4 18.1 23.2 16.2 11 28.7 16.6 11.2 5.1 10.0 19.1 22.0 16.0 12 28.0 18.0 12.811.6 14.3 16.8 20.2 16.0 13 28.8 25.2 12.510.3 16.4 19.1 20.4 16.2 14 28.0 19.1 13.411.1 15.5 19.7 25.6 13.0 25.8 13.2 10.1 8.7 13.0 16.1 21.7 19.3 16 27.7 23.4 14.110.8 12.4 17.0 23.7 23.9 17 28.3 18.3 11.1 8.2 13.4 16.2 24.8 18.6 18 28.8 20.9 11.4 7.8 13.0 16.6 19.2 19.2 19 28.0 22.3 14.3 6.9 14.5 19.3 21.3 24.8 Table I continued Rat Time (hours' 0.0 0.5 1.0 2.0 3.0 5.0 6.0 24.0 20 27.1 16.0 11.2 7.7 13.6 17.9 20.4 28.0 21 26.8 17.5 10.1 5.7 14.3 19.2 27.7 28.0 22 27.3 14.2 12.5 6.9 11.4 23.3 27.8 27.9 Table II, Plasma insulin levels (~U/ml):
Rat 23 24 25 26 27 Time 5 15.2 12.1 16.7 14.2 13.8 mins 7 48.3 37.5 43.3 40.6 42.9 54.7 52.8 55.6 49.5 52.8 52.6 45.2 50.5 48.7 51.1 32.0 35.3 31.0 28.7 30.2 19.2 12.2 12.5 11.8 12.3 7.7 5.8 4.0 5.1 4.1 3.5 4.4 2.1 3.2 3.3 120 2.0 2.8 1.6 1.9 2.5 Table I shows that the orally administered insulin formulation has a metabolic effect on the blood glucose levels. It is clear that within 2 hours the blood glucose level reached a normal level (7 mmol/L) from an initial level of about 30 mmol/L. The onset of action was very fast, i.e. about 10 minutes after administration and m~x;mum serum level was achieved within 15 minutes and the effect lasted for 24 hours with a single dose of the oral insulin formulation.
Example II
For comparative purposes, a composition containing only one absorption enhancer was tested. A composition comprising 0.3 g sodium cholate, 0.2 soyabean trypsin inhibitor, 16 IU insulin, 8.4 ml saline solution and 0.2 g polyethylene glycol in 10 ml of distilled water was prepared. The composition is not within the scope .

of the present invention. The procedure of Example I
was followed and 1 ml aliquots were gavaged to rats.
10 ~l blood samples were drawn from each rat at 0 mins, 30 mins, 1, 2, 3, 4 and 24 hours after administration of the dose, for glucose level analysis. The results are shown in Table III.

Table III, Changes in blood glucose levels (mmol/L):
Rat Time (hours) 0.0 0.5 1.0 2.0 3.0 4.0 24.0 31 30.3 28.5 26.0 28.0 20.1 15.8 28.0 32 28.5 28.0 27.1 25.0 28.0 21.7 28.0 33 29.0 28.0 26.0 28.1 28.0 24.2 28.2 34 28.7 28.1 26.5 23.0 26.9 23.0 29.6 27.8 28.2 28.0 24.1 21.3 18.2 30.3 36 28.9 27.5 28.0 20.5 21.5 23.8 28.0 37 28.1 28.2 26.5 24.0 27.5 28.0 29.2 38 28.9 26.8 24.0 19.0 20.2 21.8 29.7 Table III shows that the orally administered insulin formulation which contains only one absorption enhancer, i.e sodium cholate, has very little metabolic effect on the blood glucose levels.
Example III
0.25 g of oleic acid, 0.25 g of linoleic acid, 0.1 g sodium lauryl sulphate and 0.25 ml of Tween~ 80 polyoxyethylene sorbitan ester were dissolved in a small quantity of cold (4~C) distilled water which contained 0.2% gelatin and 0.9% sodium chloride. Dissolution was effected with rapid stirring, and the volume topped up to 9 ml with distilled water. To this clear suspension was added 1 ml of Novolin-R (trade mark) fast acting insulin. The mixture was stirred continuously to give a homogenous solution.
Aliquots of the solution were administered to diabetic (urine glucose positive) male wistar BB strain rats as in Example I and blood glucose levels were determined at intervals of 0 min, 30 min, 1, 2, 3 and 5 hours after administration of the dose. The results are shown in Table IV.

Table IV, Changes in blood glucose levels (mmol/L):
Rat Time (hours) 0.0 0.5 1.0 2.0 3.0 5.0 41 28.5 21.8 11.8 8.9 13.2 12.6 42 29.0 18.3 18.0 13.2 12.8 14.8 43 28.7 12.0 8.7 9.8 11.4 13.4 44 30.1 18.1 10.4 8.8 13.8 25.6 Example IV
The experiment of Example III was repeated, except that the composition comprised 0.25 g of disodium EDTA, 0.25 g of sodium salicylate and 0.25 g of polyoxyethylene 9-lauryl ether dissolved in a small quantity of cold (4~C) distilled water which contained 0.2% gelatin and 0.9~ sodium chloride. The results are shown in Table V below.
Table V, Chanqes in blood glucose levels (mmol/L):

Rat Time (hours) 0.0 0.5 1.0 2.0 3.0 5.0 51 28.6 28.1 17.9 15.2 13.8 25.0 52 29.8 16.1 13.0 11.8 12.2 18.2 53 29.6 17.0 10.3 10.1 15.1 23.7 54 30.1 11.4 11.5 9.9 11.8 15.3 Example V
The experiment of Example III was repeated, except that the composition comprised 0.1 g of monoolein, 0.25 g of deoxycholate and 0.25 g of polyoxyethylene 9-CA 022l0996 l997-07-l8 lauryl ether dissolved in a small quantity of cold (4~C) distilled water which contained 0. 2% gelatin and 0.9%
sodium chloride. The results are shown in Table VI.

Table VI, Changes in blood glucose levels (mmol/L):
Rat Time (hours) 0.0 0.5 1.0 2.0 3.0 5.0 61 28.1 14.5 11.2 10.2 10.3 14.3 62 28.0 26.0 21.9 12.2 8.0 15.8 0 63 29.8 24.3 20.0 15.4 12.8 14.2 64 29.5 18.4 14.2 10.2 11.0 17.0 Examples III, IV and V all show the metabolic effect on the blood glucose levels of the orally administered insulin.

Claims (16)

CLAIMS:
1. A liquid pharmaceutical agent formulation suitable for oral or nasal delivery comprising a proteinic pharmaceutical agent, water and at least two absorption enhancing compounds selected from the group consisting of sodium salicylate, sodium lauryl sulphate, disodium ethylenediaminetetraacetic acid (disodium EDTA), oleic acid, linoleic acid, monoolein, lecithin, lysolecithin, deoxycholate, sodium deoxycholate, chenodeoxycholate, taurodeoxycholate, glycochenodeoxycholate, polyoxyethylene X-lauryl ether wherein X is from 9 to 20, sodium tauro-24, 25-dihydrofusidate, polyoxyethylene ether, polyoxyethylene sorbitan esters, p-t-octylphenoxypolyoxyethylene, -lauryl-.beta.-D-maltopyranoside, 1-dodecylazacycloheptane-2-azone and phospholipids, wherein the amount of each of the absorption enhancing compounds is present in a concentration of from 1 to 10 wt./wt.% of the total formulation.
2. A liquid pharmaceutical agent formulation according to Claim 1 wherein the concentration of each of the absorption enhancing compounds is from 1 to 5 wt./wt.%.
3. A liquid pharmaceutical agent formulation according to claim 2 wherein the concentration of each of the absorption enhancing compounds is from 1.5 wt./wt.% to 3.5 wt./wt.%
4. A liquid pharmaceutical agent formulation according to Claim 2 or 3 wherein the absorption enhancing compounds are selected from the group consisting of i) a combination of deoxycholate, chenodeoxycholate, and polyoxyethylene 9-lauryl ether; ii) a combination of sodium salicylate, deoxycholate, chenodeoxycholate, and polyoxyethylene 9-lauryl ether; iii) a combination of sodium deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and monoolein; iv) a combination of deoxycholate, chenodeoxycholate and sodium salicylate;
v) a combination of deoxycholate, sodium salicylate and sodium lauryl sulphate; vi) a combination of deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and sodium tauro-24, 25-dihydrofusidate;
vii) a combination of sodium deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and sodium tauro-24, 25-dihydrofusidate; viii) a combination of deoxycholate, chenodeoxycholate, taurodeoxycholate, polyoxyethylene 9-lauryl ether and monoolein; ix) a combination of chenodeoxycholate,glycochenodeoxycholate, polyoxyethylene 9-lauryl ether and sodium tauro-24, 25-dihydrofusidate; x) a combination of chenodeoxycholate, sodium lauryl sulphate and disodium EDTA; xi) a combination of deoxycholate, chenodeoxycholate, polyoxyethylene 9-lauryl ether and disodium EDTA; xii) a combination of sodium salicylate, disodium EDTA and polyoxyethylene 9-lauryl ether; xiii) a combination of monoolein, oleic acid and polyoxyethylene sorbitan ester; xiv) a combination of monoolein, oleic acid, polyoxyethylene sorbitan ester and sodium lauryl sulphate; xv) and a combination of linoleic acid, monoolein and sodium salicylate.
5. A liquid pharmaceutical agent formulation according to Claim 4 wherein the absorption enhancing compound compositions are selected from the group consisting of i) a combination of deoxycholate, chenodeoxycholate and sodium salicylate, ii) a combination of sodium salicylate, deoxycholate and sodium lauryl sulphate, iii) a combination of chenodeoxycholate, sodium lauryl sulphate and disodium EDTA and iv) a combination of monoolein, oleic acid and polyoxyethylene sorbitan ester, and v) a combination of deoxycholate, chenodeoxycholate and polyoxyethylene 9-lauryl ether.
6. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3, or 4 wherein the formulation contains at least one protective polymer for slow release of the pharmaceutical agent.
7. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3, or 4 wherein the formulation contains at least one protective polymer selected from the group consisting of polyvinyl alcohol, polyethylene glycol and gelatin.
8. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3 or 4 wherein the formulation contains an antioxidant.
9. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3 or 4 wherein the formulation contains an antioxidant selected from the group consisting of tocopherol, deteroxime mesylate, methyl paraben and ascorbic acid.
10. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3 or 4 wherein the formulation contains at least one protease inhibitor.
11. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3 or 4 wherein the formulation contains a protease inhibitor selected from the group consisting of bacitracin, soyabean trypsin and aprotinin in a concentration of from 1 to 3 wt./wt.%.
12. A liquid pharmaceutical agent formulation according to any one of Claims 1, 2, 3 or 4 wherein the pharmaceutical agent is selected from the group consisting of insulin, heparin, hirugen, hirulos, huridine, mumps, measles and rubella vaccine, typhoid vaccine, hepatitis A vaccine, hepatitis B vaccine, herpes simplex virus, bacterial toxoids, cholera toxin B-subunit, influenza vaccine virus, whooping cough vaccine virus, vaccinia virus, adenovirus, canary pox, polio vaccine virus, malaria vaccine virus, bacillus calmette geurin (BCG), klebsiella pneumoniae, HIV
envelop glycoproteins, bovine somatropine, estrogens, androgens, insulin growth factors, interleukin-I, interleukin-II and cytokins.
13. A liquid pharmaceutical agent formulation according to Claim 4 wherein the pharmaceutical agent is insulin.
14. A liquid pharmaceutical agent formulation according to Claim 13 wherein the formulation also contains at least one inorganic salt which opens channels in the gastrointestinal tract, at least one protective polymer and at least one protease inhibitor.
15. A liquid pharmaceutical agent formulation according to Claim 14 wherein the inorganic salt is selected from the group consisting of sodium chloride, potassium chloride, calcium chloride, zinc chloride and sodium bicarbonate, the protective polymer is selected from the group consisting of polyvinyl alcohol, polyethylene glycol and gelatin, and the protease inhibitor is selected from the group consisting of bacitracin, soyabean trypsin and aprotinin.
16. A liquid pharmaceutical agent formulation according to Claim 15 which also contains an antioxidant.
CA002210996A 1995-05-16 1996-05-16 Liquid formulations for proteinic pharmaceuticals Expired - Fee Related CA2210996C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/442,358 US5653987A (en) 1995-05-16 1995-05-16 Liquid formulations for proteinic pharmaceuticals
US08/442,358 1995-05-16
PCT/CA1996/000305 WO1996036352A1 (en) 1995-05-16 1996-05-16 Liquid formulations for proteinic pharmaceuticals comprising at least 2 absorption enhancers

Publications (2)

Publication Number Publication Date
CA2210996A1 CA2210996A1 (en) 1996-11-21
CA2210996C true CA2210996C (en) 2001-04-03

Family

ID=23756510

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002210996A Expired - Fee Related CA2210996C (en) 1995-05-16 1996-05-16 Liquid formulations for proteinic pharmaceuticals

Country Status (5)

Country Link
US (1) US5653987A (en)
EP (1) EP0813421A1 (en)
AU (1) AU5642396A (en)
CA (1) CA2210996C (en)
WO (1) WO1996036352A1 (en)

Families Citing this family (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050032844A1 (en) * 1996-07-17 2005-02-10 Nicogen, Inc. Methods for regulating nicotine metabolism
US6017946A (en) * 1997-10-08 2000-01-25 Posner; Robert Serotonin containing formulation for oral administration and method of use
US6908631B1 (en) * 1997-12-01 2005-06-21 Nicogen, Inc. Therapeutic and diagnostic methods dependent on CYP2A enzymes
US6221378B1 (en) * 1998-02-10 2001-04-24 Generex Pharmaceuticals Incorporated Mixed micellar delivery system and method of preparation
US7070799B1 (en) * 1998-02-10 2006-07-04 Generex Pharmaceuticals, Inc. Method for administering insulin to the buccal region
US6017545A (en) * 1998-02-10 2000-01-25 Modi; Pankaj Mixed micellar delivery system and method of preparation
US6350458B1 (en) 1998-02-10 2002-02-26 Generex Pharmaceuticals Incorporated Mixed micellar drug deliver system and method of preparation
US7303768B2 (en) 1998-07-24 2007-12-04 Seo Hong Yoo Preparation of aqueous clear solution dosage forms with bile acids
US7772220B2 (en) * 2004-10-15 2010-08-10 Seo Hong Yoo Methods and compositions for reducing toxicity of a pharmaceutical compound
US20050158408A1 (en) * 1998-07-24 2005-07-21 Yoo Seo H. Dried forms of aqueous solubilized bile acid dosage formulation: preparation and uses thereof
EP1113785B1 (en) * 1998-07-24 2005-04-13 Seo Hong Yoo Clear aqueous solutions of bile acids
US6703381B1 (en) 1998-08-14 2004-03-09 Nobex Corporation Methods for delivery therapeutic compounds across the blood-brain barrier
TW570805B (en) 1998-09-01 2004-01-11 Hoffmann La Roche Water-soluble pharmaceutical composition in an ionic complex
US6193997B1 (en) * 1998-09-27 2001-02-27 Generex Pharmaceuticals Inc. Proteinic drug delivery system using membrane mimetics
US6290987B1 (en) * 1998-09-27 2001-09-18 Generex Pharmaceuticals, Inc. Mixed liposome pharmaceutical formulation with amphiphiles and phospholipids
EP1137431A1 (en) * 1998-12-04 2001-10-04 Provalis UK Limited Pharmaceutical compositions containing insulin
US6312665B1 (en) * 1998-12-21 2001-11-06 Generex Pharmaceuticals Incorporated Aerosol formulations for buccal and pulmonary application
US6451286B1 (en) 1998-12-21 2002-09-17 Generex Pharmaceuticals Incorporated Pharmaceutical compositions for buccal and pulmonary administration comprising an alkali metal alkyl sulfate and at least three micelle-forming compounds
EP1338272A1 (en) * 1998-12-21 2003-08-27 Generex Pharmaceuticals Inc. Aerosol formulations for buccal and pulmonary application comprising chenodeoxycholate or deoxycholate
US6294153B1 (en) * 1998-12-21 2001-09-25 Generex Pharmaceuticals, Inc. Aerosol pharmaceutical formulation for pulmonary and nasal delivery
US7087215B2 (en) * 1998-12-21 2006-08-08 Generex Pharmaceuticals Incorporated Methods of administering and enhancing absorption of pharmaceutical agents
US6436367B1 (en) * 1998-12-21 2002-08-20 Generex Pharmaceuticals Inc. Aerosol formulations for buccal and pulmonary application
US6849263B2 (en) * 1998-12-21 2005-02-01 Generex Pharmaceutical Incorporated Pharmaceutical compositions for buccal delivery of pain relief medications
AU2006200276B2 (en) * 1998-12-21 2007-11-29 Generex Pharmaceuticals Inc. Micellar pharmaceutical compositions for buccal and pulmonary application
US6271200B1 (en) 1998-12-21 2001-08-07 Generex Pharmaceuticals Inc. Proteinic drug delivery system using aerosolized membrane-mimetic amphiphiles
US6375975B1 (en) * 1998-12-21 2002-04-23 Generex Pharmaceuticals Incorporated Pharmaceutical compositions for buccal and pulmonary application
SI1031347T1 (en) * 1999-01-27 2002-10-31 Idea Ag Transnasal transport/immunisation with highly adaptable carriers
SI1031346T1 (en) 1999-01-27 2002-08-31 Idea Ag Noninvasive vaccination through the skin
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
US6761903B2 (en) 1999-06-30 2004-07-13 Lipocine, Inc. Clear oil-containing pharmaceutical compositions containing a therapeutic agent
US6350432B1 (en) 1999-03-19 2002-02-26 Generex Pharmaceuticals Incorporated Pressurized container having an aerosolized pharmaceutical composition
US6383471B1 (en) 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
US6410255B1 (en) * 1999-05-05 2002-06-25 Aurora Biosciences Corporation Optical probes and assays
ATE370928T1 (en) 1999-05-14 2007-09-15 Nereus Pharmaceuticals Inc INTERLEUKIN-1 AND TUMOR NECROSIS FACTOR ALPHA MODULATORS, SYNTHESIS OF THESE MODULATORS AND METHODS OF APPLYING THESE MODULATORS
US6309663B1 (en) 1999-08-17 2001-10-30 Lipocine Inc. Triglyceride-free compositions and methods for enhanced absorption of hydrophilic therapeutic agents
US6458383B2 (en) 1999-08-17 2002-10-01 Lipocine, Inc. Pharmaceutical dosage form for oral administration of hydrophilic drugs, particularly low molecular weight heparin
US6982281B1 (en) * 2000-11-17 2006-01-03 Lipocine Inc Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs
JP4763197B2 (en) * 1999-09-24 2011-08-31 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム New vaccine
MXPA02003067A (en) * 1999-09-24 2002-09-30 Smithkline Beecham Biolog Use of combination of polyoxyethylene sorbitan ester and octoxynol as adjuvant and its use in vaccines.
AR025749A1 (en) * 1999-09-24 2002-12-11 Smithkline Beecham Biolog VACCINES
YU31902A (en) * 1999-10-28 2004-12-31 Institut Neftekhimicheskogo Sinteza Imeni A.V.Topchieva Rossiiskoi Akademii Nauk Polypeptide composition
US7732404B2 (en) 1999-12-30 2010-06-08 Dexcel Ltd Pro-nanodispersion for the delivery of cyclosporin
KR100848344B1 (en) * 2000-02-04 2008-07-25 유서홍 Preparation of aqueous clear solution dosage forms with bile acids
WO2001062919A1 (en) * 2000-02-23 2001-08-30 Aurora Biosciences Corporation Modified fluorescent proteins
WO2001072278A2 (en) * 2000-03-30 2001-10-04 Generex Pharmaceuticals Inc. Method for administering insulin to the buccal region
US20080319025A1 (en) * 2000-06-01 2008-12-25 Nicogen, Inc. Therapeutic and Diagnostic Methods Dependent on CYP2A Enzymes
US7144723B2 (en) * 2000-11-16 2006-12-05 The Regents Of The University Of California Marine actinomycete taxon for drug and fermentation product discovery
CN100453079C (en) * 2000-11-24 2009-01-21 润风美有限公司 Water-soluble liquid internal medicine
AU3664102A (en) * 2000-12-01 2002-06-11 Battelle Memorial Institute Method for stabilizing biomolecules in liquid formulations
US7060675B2 (en) 2001-02-15 2006-06-13 Nobex Corporation Methods of treating diabetes mellitus
US20020141970A1 (en) * 2001-03-05 2002-10-03 Pettit Dean K. Stable aqueous solutions of granulocyte macrophage colony-stimulating factor
US20030031757A1 (en) * 2001-08-03 2003-02-13 Kraft Food Holdings, Inc. Stable and bioavailable iron fortified beverages
DK1429731T3 (en) * 2001-09-19 2007-05-14 Elan Pharma Int Ltd Nanoparticle formulations containing insulin
US7176232B2 (en) 2002-06-24 2007-02-13 The Regents Of The University Of California Salinosporamides and methods for use thereof
WO2004034970A2 (en) * 2002-09-27 2004-04-29 Nereus Pharmaceuticals, Inc. Macrocyclic lactams
GB0308734D0 (en) * 2003-04-15 2003-05-21 Axcess Ltd Uptake of macromolecules
GB0308732D0 (en) * 2003-04-15 2003-05-21 Axcess Ltd Absorption enhancers
NZ544588A (en) * 2003-06-20 2010-06-25 Nereus Pharmaceuticals Inc Use of salinosporamide A and analogs thereof for the treatment of cancer, inflammation and infectious diseases
CN1823070A (en) * 2003-06-20 2006-08-23 加利福尼亚大学董事会 Salinosporamides and methods for use thereof
US7145125B2 (en) 2003-06-23 2006-12-05 Advanced Optical Technologies, Llc Integrating chamber cone light using LED sources
US8211448B2 (en) 2003-07-07 2012-07-03 Nares Ab Microemulsions and its use for preventing airway diseases
PL1648412T3 (en) * 2003-07-07 2008-04-30 Nares Ab Microemulsions and its use for preventing airway diseases
US7727752B2 (en) 2003-07-29 2010-06-01 Life Technologies Corporation Kinase and phosphatase assays
US20050043253A1 (en) * 2003-08-19 2005-02-24 Cook Bradley R. Use of wound healing compositions for prevention of infections and allergies
US20050058702A1 (en) * 2003-09-17 2005-03-17 Ben-Sasson Shmuel A. Compositions capable of facilitating penetration across a biological barrier
WO2005065185A2 (en) * 2003-12-24 2005-07-21 Collegium Pharmaceuticals, Inc. Temperature-stable formulations, and methods of development thereof
US7658721B2 (en) * 2004-01-16 2010-02-09 Biodel Inc. Sublingual drug delivery device
AU2005206560A1 (en) * 2004-01-23 2005-08-04 Nereus Pharmaceuticals, Inc. Bis-indole pyrroles useful as antimicrobials agents
ES2398838T3 (en) * 2004-03-12 2013-03-22 Biodel, Inc. Fast-acting drug delivery compositions
US20080248999A1 (en) * 2007-04-04 2008-10-09 Biodel Inc. Amylin formulations
US20080090753A1 (en) 2004-03-12 2008-04-17 Biodel, Inc. Rapid Acting Injectable Insulin Compositions
US20080096800A1 (en) * 2004-03-12 2008-04-24 Biodel, Inc. Rapid mucosal gel or film insulin compositions
AU2005283141B2 (en) * 2004-04-30 2012-05-10 Nereus Pharmaceuticals, Inc. (3.2.0) heterocyclic compounds and methods of using the same
US7579371B2 (en) 2004-04-30 2009-08-25 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US8173627B2 (en) * 2004-08-30 2012-05-08 Seo Hong Yoo Neuroprotective effect of solubilized UDCA in focal ischemic model
AU2005295541B2 (en) * 2004-10-15 2011-02-17 Seo Hong Yoo Methods and compositions for reducing toxicity of a pharmaceutical compound
WO2006050165A2 (en) * 2004-11-01 2006-05-11 Seo Hong Yoo Methods and compositions for reducing neurodegeneration in amyotrophic lateral sclerosis
PL1830838T3 (en) * 2004-12-03 2013-07-31 Dana Farber Cancer Inst Inc Compositions and methods for treating neoplastic diseases
WO2007006320A1 (en) * 2005-07-12 2007-01-18 Sherine Hassan Abbas Helmy Drinkable oral insulin liquid and capsules
AU2006276274B2 (en) 2005-07-21 2012-03-29 Nereus Pharmaceuticals, Inc. Interleukin-1 and tumor necrosis factor-a modulators; syntheses of such modulators and methods of using such modulators
US7713929B2 (en) * 2006-04-12 2010-05-11 Biodel Inc. Rapid acting and long acting insulin combination formulations
US8084420B2 (en) * 2005-09-29 2011-12-27 Biodel Inc. Rapid acting and long acting insulin combination formulations
MX2008013165A (en) * 2006-04-12 2009-01-29 Biodel Inc Rapid acting and long acting insulin combination formulations.
US20100160442A1 (en) * 2006-07-18 2010-06-24 Ossovskaya Valeria S Formulations for cancer treatment
CA2662607A1 (en) * 2006-09-08 2008-03-13 Kaneka Corporation Composition comprising reduced coenzyme q10 and lysolecithin
US7824698B2 (en) * 2007-02-02 2010-11-02 Nereus Pharmaceuticals, Inc. Lyophilized formulations of Salinosporamide A
US20090017167A1 (en) * 2007-07-11 2009-01-15 Herbalife International Inc. Mixture and beverage made therefrom for protecting cellular hydration
US8394816B2 (en) * 2007-12-07 2013-03-12 Irene Ghobrial Methods of using [3.2.0] heterocyclic compounds and analogs thereof in treating Waldenstrom's Macroglobulinemia
WO2009089181A1 (en) * 2008-01-04 2009-07-16 Blodel, Inc. Insulin formulations for insulin release as a function of tissue glucose levels
EP2276765A4 (en) * 2008-05-12 2011-10-19 Nereus Pharmaceuticals Inc Salinosporamide derivatives as proteasome inhibitors
US11304960B2 (en) 2009-01-08 2022-04-19 Chandrashekar Giliyar Steroidal compositions
US9060927B2 (en) * 2009-03-03 2015-06-23 Biodel Inc. Insulin formulations for rapid uptake
HUP0900482A2 (en) 2009-08-03 2011-03-28 Cera Med Kft Pharmaceutical formulation for oral administration
US20110142889A1 (en) * 2009-12-16 2011-06-16 Nod Pharmaceuticals, Inc. Compositions and methods for oral drug delivery
US9358241B2 (en) 2010-11-30 2016-06-07 Lipocine Inc. High-strength testosterone undecanoate compositions
US9034858B2 (en) 2010-11-30 2015-05-19 Lipocine Inc. High-strength testosterone undecanoate compositions
US20180153904A1 (en) 2010-11-30 2018-06-07 Lipocine Inc. High-strength testosterone undecanoate compositions
US20120148675A1 (en) 2010-12-10 2012-06-14 Basawaraj Chickmath Testosterone undecanoate compositions
WO2016033556A1 (en) 2014-08-28 2016-03-03 Lipocine Inc. BIOAVAILABLE SOLID STATE (17-β)-HYDROXY-4-ANDROSTEN-3-ONE ESTERS
WO2016033549A2 (en) 2014-08-28 2016-03-03 Lipocine Inc. (17-ß)-3-OXOANDROST-4-EN-17-YL TRIDECANOATE COMPOSITIONS AND METHODS OF THEIR PREPARATION AND USE
CA2959965A1 (en) 2014-09-02 2016-03-10 Bhupinder Singh Pharmaceutical formulations comprising tetrahydrocurcumin and lipids
EP3204120B1 (en) 2014-10-08 2021-05-26 Pacific Northwest Research Institute Methods and compositions for increasing the potency of antifungal agents
CA3078723A1 (en) 2016-11-28 2018-05-31 Nachiappan Chidambaram Oral testosterone undecanoate therapy
EP3773612A4 (en) 2018-03-28 2022-01-12 Herbalife International of America, Inc. Acetylation of polysaccharides
IL294979A (en) * 2020-01-23 2022-09-01 Axcess Uk Ltd Cellular uptake

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4900730A (en) * 1981-01-14 1990-02-13 Toyo Jozo Co., Ltd. Preparation which promotes the absorption of peptides
JPS61267528A (en) * 1984-11-26 1986-11-27 Yamanouchi Pharmaceut Co Ltd Transnasal calcitonin agent containing absorbefacient
IL78425A (en) * 1985-04-15 1991-05-12 Lilly Co Eli Intranasal formulation containing insulin
NZ222907A (en) * 1986-12-16 1990-08-28 Novo Industri As Preparation for intranasal administration containing a phospholipid absorption enhancing system
US4789660A (en) * 1987-09-10 1988-12-06 American Home Products Corporation Insulin administration using methyl and propyl paraben
AT392906B (en) * 1987-10-08 1991-07-10 Hoffmann La Roche Pharmaceutical products for oral administration
GB9012663D0 (en) * 1990-06-07 1990-08-01 Erba Carlo Spa Galenic formulations containing cyclodextrins
US5206219A (en) * 1991-11-25 1993-04-27 Applied Analytical Industries, Inc. Oral compositions of proteinaceous medicaments
CA2094217A1 (en) * 1992-04-17 1993-10-18 Yasutaka Igari Transmucosal therapeutic composition
US5346701A (en) * 1993-02-22 1994-09-13 Theratech, Inc. Transmucosal delivery of macromolecular drugs
US5447729A (en) * 1994-04-07 1995-09-05 Pharmavene, Inc. Multilamellar drug delivery systems

Also Published As

Publication number Publication date
CA2210996A1 (en) 1996-11-21
WO1996036352A1 (en) 1996-11-21
EP0813421A1 (en) 1997-12-29
US5653987A (en) 1997-08-05
AU5642396A (en) 1996-11-29

Similar Documents

Publication Publication Date Title
CA2210996C (en) Liquid formulations for proteinic pharmaceuticals
US4548922A (en) Drug administration
KR100767097B1 (en) Stable topical drug delivery compositions
JP4499282B2 (en) Mixed micellar drug transfer system and preparation method
EP0521994B1 (en) Pharmaceutical formulations
EP0128831B1 (en) Drug administration
US8273711B2 (en) Topical drug delivery using phosphatidylcholine
US6951655B2 (en) Pro-micelle pharmaceutical compositions
EP0219076B1 (en) Sustained release composition
US5626873A (en) Emulsions
EP1326644B1 (en) Absorption enhancers
IE61110B1 (en) Nasal formulations and a process for preparation thereof
Ziv et al. Oral administration of insulin in solid form to nondiabetic and diabetic dogs
US20100203105A1 (en) Method for administering insulin to the buccal region
EP0177342A2 (en) Oral formulation of therapeutic proteins
EP0351651B1 (en) Insulin preparation
WO2001072278A2 (en) Method for administering insulin to the buccal region
EP0213552A2 (en) Oily suspension for intrarectal infusion
CZ192099A3 (en) Topic preparation for introducing peptide pharmaceuticals into vivid organisms
EP0535827A1 (en) Calcitonin suspensions for oral administration
Gelb Effect of hypophysectomy, thyroidectomy, adrenalectomy and alloxan diabetes on incorporation of fatty acids into esters by the small intestine in vitro
JP2002069000A (en) Oral medicine for treating diabetes mellitus
CN1163573A (en) Pharmaceutical compositions containing bile salt and buffer for increased bioavailability of active compound
SI8712284A (en) Preparations for nasal application and procedures for their production.

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20140516