US20050054665A1 - Treatment for CD5+ B cell lymphoma - Google Patents

Treatment for CD5+ B cell lymphoma Download PDF

Info

Publication number
US20050054665A1
US20050054665A1 US10/933,594 US93359404A US2005054665A1 US 20050054665 A1 US20050054665 A1 US 20050054665A1 US 93359404 A US93359404 A US 93359404A US 2005054665 A1 US2005054665 A1 US 2005054665A1
Authority
US
United States
Prior art keywords
cell lymphoma
cells
lymphoma cells
cell
irm
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
US10/933,594
Inventor
Richard Miller
David Spaner
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.)
Coley Pharmaceutical Group Inc
Original Assignee
3M Innovative Properties Co
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 3M Innovative Properties Co filed Critical 3M Innovative Properties Co
Priority to US10/933,594 priority Critical patent/US20050054665A1/en
Assigned to 3M INNOVATIVE PROPERTIES COMPANY reassignment 3M INNOVATIVE PROPERTIES COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MILLER, RICHARD L., SPANER, DAVID E.
Publication of US20050054665A1 publication Critical patent/US20050054665A1/en
Assigned to COLEY PHARMACEUTICAL GROUP, INC. reassignment COLEY PHARMACEUTICAL GROUP, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: 3M COMPANY; 3M INNOVATIVE PROPERTIES COMPANY
Priority to US12/173,719 priority patent/US7923429B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid

Definitions

  • the peripheral B cell neoplasm chronic lymphoid leukemia/small lymphocytic lymphoma represents the most common lymphoid leukemia. As the name implies, presentation can be as either leukemia or lymphoma. However, the two presentations of this neoplasm, chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL), are morphologically, phenotypically, and genotypically indistinguishable, differing only in the degree of peripheral blood lymphocytosis.
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • CLL is the most common leukemia of adults in the Western world.
  • the peripheral blood contains small, round lymphocytes with scant cytoplasm. Involvement of the bone marrow is observed in all cases of CLL and most cases of SLL, taking the form of interstitial infiltrates or nonparatubular aggregates of small lymphocytes.
  • the tumor cells in CLL and SLL express the pan B cell markers CD29 and CD20.
  • CD5-a T cell marker that is expressed only on a small subset of normal B cells—is present on the tumor cells.
  • the immunophenotype of CLL cells is unique.
  • CLL cells co-express the B lymphocyte lineage marker CD19 and the T lymphocyte marker CD5.
  • CLL cells also exhibit a characteristic level of expression of immunoglobulin receptor.
  • Tumor cells typically also have low-level surface expression of Ig heavy chain, with either kappa or lambda light chains.
  • CLL is a clonal malignancy of B lymphocytes.
  • the disease is usually indolent, with slowly progressive accumulation of long-lived small lymphocytes that are immunoincompetent and respond poorly to antigenic stimulation.
  • CLL is incurable with conventional cytotoxic chemotherapy (Cheson et al., Blood 1996; 87:4990-4997; and Keating et al., Blood 1993; 81:2878-2884).
  • the hallmark of CLL is isolated lymphocytosis.
  • the white blood cell count is usually greater than 20,000 ⁇ L and may be markedly elevated to several hundred thousand.
  • the diagnostic requirement for CLL is an absolute lymphocyte count of greater than 4000/mm 3 .
  • CLL is manifested clinically by immunosuppression, bone marrow failure, and organ infiltration with lymphocytes. Immunodeficiency is also related to inadequate antibody production by the abnormal B cells. With advanced disease, CLL may cause damage by direct tissue infiltration.
  • patients may develop cutaneous lymphoma deposits—erythomatous lesions on the skin.
  • the lesions may contain an atypical lymphoid dermal infiltrate of small, round B cells.
  • IRMs small molecule immune response modifiers
  • CD5 + B cell lymphomas such as, for example, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), mantle cell lymphoma, or splenic lymphoma with villous lymphocytes.
  • the invention provides a method of treating a CD5 + B cell lymphoma.
  • the method includes administering to a subject an IRM compound in an amount effective to ameliorate at least one symptom or clinical sign of the CD5 + B cell lymphoma.
  • administering the IRM compound may result in at least a 50% decrease in peripheral blood lymphocytes, lymphadenopathy, or splenomegaly for at least two months.
  • administering the IRM compound can inhibit or even prevent the development of progressive disease, wherein progressive disease is at least a 50% increase in circulating lymphocytes or a progression to a more aggressive histology.
  • administering the IRM compound can resolve nodular, erythematous lesions associated with CD5 + B cell lymphoma.
  • the invention provides a method of increasing the expression of at least one cell surface molecule of CD5 + B cell lymphoma cells.
  • the method includes contacting the CD5 + B cell lymphoma cells with at least one IRM effective for increasing the expression of at least one cell surface molecule of the CD5 + B cell lymphoma cells.
  • the cell surface molecule may be a costimulatory molecule.
  • the present invention also provides a method of stimulating CD5 + B cell lymphoma cells to produce a cytokine by contacting the CD5 + B cell lymphoma cells with an IRM effective for inducing production of a cytokine above a level produced by the CD5 + B cell lymphoma cells not contacted by the IRM.
  • the cytokine may be IL-1 ⁇ , IL-6, IL-8, IL-10, IL-12, TNF- ⁇ , GM-CSF, or combinations thereof.
  • the invention provides a method of increasing proliferation of CD5 + B cell lymphoma-specific cytotoxic T cells.
  • the method includes contacting CD5 + B cell lymphoma cells with an IRM effective to increase the expression of at least one costimulatory molecule on the surface of CD5 + B cell lymphoma cells, and then contacting CD8 + T cells with the CD5 + B cell lymphoma cells, thereby activating the CD8 + T cells; wherein the activated T cells are CD5 + B cell lymphoma-specific cytotoxic T cells and demonstrate increased proliferation compared to T cells contacted with CD5 + B cell lymphoma cells that have not been contacted with an IRM.
  • the CD8 + T cells are CD5 + B cell lymphoma cell-specific. In other embodiments, the CD8 + T cells are naive.
  • the IRM compound may be administered to a subject diagnosed as having a CD5 + B cell lymphoma so that the activated CD5 + B cell lymphoma-specific cytotoxic T cells are autologous CD5 + B cell lymphoma-specific cytotoxic T cells.
  • the CD5 + B cell lymphoma cells may be further contacted with one or more additional immunomodulating agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • additional immunomodulating agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • the present invention also provides a method of increasing the killing of CD5 + B cell lymphoma cells by cytotoxic T cells.
  • the method includes contacting CD5 + B cell lymphoma cells with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of the CD5 + lymphoma cells, and then contacting CD8 + T cells with the CD5 + B cell lymphoma cells, thereby activating the CD8 + T cells; wherein the activated CD8 + T cells are CD5 + B cell lymphoma-specific cytotoxic T cells and demonstrate increased killing of CD5 + B cell lymphoma cells compared to T cells contacted with CD5 + B cell lymphoma cells that have not been contacted with an IRM.
  • the CD8 + T cells are CD5 + B cell lymphoma cell-specific. In other embodiments, the CD8 + T cells are naive.
  • the IRM compound may be administered to a subject diagnosed as having a CD5 + B cell lymphoma so that the activated CD5 + B cell lymphoma-specific cytotoxic T cells are autologous CD5 + B cell lymphoma-specific cytotoxic T cells.
  • the CD5 + B cell lymphoma cells may be further contacted with one or more additional immunomodulating agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • additional immunomodulating agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • the present invention also provides a method of treating a subject suffering from a CD5 + B cell lymphoma including administering to the subject an IRM effective to increase the expression of at least one cell surface molecule of the CD5 + B cell lymphoma cells.
  • the present invention also provides a vaccine that includes isolated CD5 + B cell lymphoma cells, or an immunologically active portion thereof, wherein the isolated CD5 + B cell lymphoma cells have been contacted with an IRM effective to increase the expression of at least one cell surface molecule of the CD5 + B cell lymphoma cells.
  • the CD5 + B cell lymphoma cells may be further contacted with one or more additional immunomodulatory agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • the present invention also provides a method of preparing a vaccine including contacting isolated CD5 + B cell lymphoma cells with an IRM effective to increase the expression of at least one molecule on the surface of the CD5 + B cell lymphoma cells. Some embodiments may include further contacting isolated CD5 + B cell lymphoma cells with one or more additional immunomodulatory agents such as, for example, IL-2 or a protein kinase C agonist. In other embodiments, the isolated CD5 + B cell lymphoma cells may be derived from a subject diagnosed as having CLL or SLL.
  • the CD5 + B cell lymphoma cells may be chronic lymphocytic leukemia (CLL) cells, small lymphocytic lymphoma cells (SLL), mantle cell lymphoma cells, splenic lymphoma with villous lymphocytes, or combinations thereof.
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma cells
  • mantle cell lymphoma cells splenic lymphoma with villous lymphocytes, or combinations thereof.
  • the IRM is a TLR7 agonist. In some embodiments, the IRM is a TLR8 agonist. In still other embodiments, the IRM compound may be an agonist of both TLR7 and TLR8.
  • the IRM may be an imidazoquinoline amine, a tetrahydroimidazoquinoline amine, an imidazopyridine amine, a 1,2-bridged imidazoquinoline amine, a 6,7-fused cycloalkylimidazopyridine amine, an imidazonaphthyridine amine, a tetrahydroimidazonaphthyridine amine, an oxazoloquinoline amine, a thiazoloquinoline amine, an oxazolopyridine amine, a thiazolopyridine amine, an oxazolonaphthyridine amine, or a thiazolonaphthyridine amine.
  • the IRM is an imidazoquinoline amine such as, for example, 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine.
  • the IRM is a tetrahydroimidazoquinoline amine such as, for example, 4-amino-2-(ethoxymethyl)- ⁇ , ⁇ -dimethyl-6,7,8,9-tetrahydro-1H-imidazo[4,5-c]quinoline-1-ethanol hydrate.
  • the IRM is a sulfonamide substituted imidazoquinoline amine such as, for example, N-[4-(4-amino-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)butyl]methanesulfonamide.
  • At least one cell surface molecule whose expression is increased may be CD20, CD22, or CD23, and the method further includes administering to the subject a therapeutic agent that has, as a target, the cell surface molecule whose expression is increased. In some embodiments, the expression of more than one cell surface molecule may be increased.
  • At least one costimulatory molecule whose expression is increased may be CD40, CD54, CD80, CD83, CD86, CD25, or CD38. In some embodiments, the expression of more than one costimulatory molecule may be increased.
  • the CD5 + B cell lymphoma cells may be contacted with an IRM in vitro.
  • the CD5 + B cell lymphoma cells may be contacted with an IRM in vivo such as, for example, in an organ, tissue, or blood of a subject.
  • FIGS. 1 a - b Enhancement of costimulatory molecule expression on CLL cells by an IRM compound.
  • FIGS. 2 A-C Effect of an IRM compound (with or without IL-2) on costimulatory molecule expression by CLL cells.
  • FIGS. 3 A-B Effect of an IRM compound on the ability of CLL cells to stimulate cytotoxic T cell proliferation.
  • FIGS. 4 A-C Effect of an IRM compound, IL-2, and PKC agonists on costimulatory molecule expression by CLL cells.
  • FIGS. 5 A-B Induction of T cell cytotoxicity against autologous CLL cells by PKC agonists, IL-2, and IRM.
  • FIGS. 6 A-B Photographs of a lymphomatous skin deposit before treatment ( FIG. 6A ) and after treatment ( FIG. 6B ) with an IRM.
  • FIG. 7 Effect of IRM1 on costimulatory molecule expression by CLL cells.
  • FIG. 8A -B Effect of IRM3 on expression of CD80 and CD83 by CLL cells.
  • FIG. 9 IRM3-Mediated changes in cell surface molecule expression in CLL cells.
  • FIG. 10 IRM3+IL-2 increases expression of CD20 on CLL cells.
  • the present invention provides methods for treating CD5 + B cell lymphomas such as, for example, chronic lymphocytic leukemia (CLL).
  • CD5 + B cell lymphomas such as, for example, chronic lymphocytic leukemia (CLL).
  • CLL chronic lymphocytic leukemia
  • CD5 + B cell lymphoma cells may be subject to T cell mediated immune recognition (see, for example, Ribera et al., Blood Cells 1987; 12:471-483; Ziegler-Heitbrock et al., Blood 1989; 73:1426-1430; Wierda et al., Blood 2000; 96:2917-2924; Gitelson et al., Clin Cancer Res.
  • the present invention demonstrates, for the first time, that contacting CD5 + B cell lymphoma cells with an immune response modifier (IRM) compound may be useful for treating CD5 + B cell lymphomas.
  • IRM compounds appear to act through basic immune system mechanisms known as toll-like receptors (TLRs) to induce selected biosynthesis of certain cytokines, chemokines and costimulatory molecules.
  • TLRs toll-like receptors
  • certain IRM compounds can selectively induce certain aspects—and/or inhibit other aspects—of the immune system.
  • IRM compounds may increase the expression of cell surface molecules of CD5 + B cell lymphoma cells, enhance the immunogenicity of CD5 + B cell lymphoma cells, and provide a new immunotherapeutic approach for the treatment of CD5 + B cell lymphomas.
  • the cell surface molecule whose expression is increased may be a costimulatory molecule.
  • Increasing the cell surface expression of costimulatory molecules may allow the CD5 + B cell lymphoma cells to become competent antigen presenting cells (APCs) capable of initiating and/or maintaining tumor-reactive T cell activity.
  • APCs competent antigen presenting cells
  • “Ameliorate” refers to any reduction in the extent, severity, frequency, and/or likelihood of a symptom or clinical sign characteristic of a particular condition.
  • CD5 + B cell lymphoma cells refers to neoplastic cells having a unique immunophenotype that includes co-expression of CD19 and CD5.
  • CD5 + B cell lymphoma cells also express the T lymphocyte marker CD5, which is typically expressed only on a small subset of normal B cells.
  • CD5 + B cell lymphoma cells include, for example, chronic lymphocytic leukemia (CLL) cells, small lymphocytic lymphoma (SLL) cells, mantle cell lymphoma cells, and splenic lymphoma with villous lymphocytes.
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • the CD5 + B cell lymphoma cells are CLL cells or SLL cells.
  • the CD5 + B cell lymphoma cells are CLL cells.
  • Cell surface molecule refers to a molecule that is expressed on the surface of a cell and may be used to determine the cell's lineage or otherwise may be used to distinguish one cell or cell type from another.
  • “Patient” or “Subject” includes, for example, animals such as, but not limited to, humans, non-human primates, rodents, dogs, cats, horses, pigs, sheep, goats, or cows.
  • “Sign” or “clinical sign” refers to an objective physical finding relating to a particular condition capable of being found by one other than the patient.
  • Symptom refers to any subjective evidence of disease or of a patient's condition.
  • a,” “an,” “the,” and “at least one” are used interchangeably and mean one or more than one.
  • IRM compounds such as, for example, agonists of TLR7 and/or TLR8 can increase the expression of a number of cell surface molecules (including, e.g., costimulatory molecules) of CD5 + B cell lymphoma cells, which can result in a more potent immune response being raised against the CD5 + B cell lymphoma cells.
  • cell surface molecules of CD5 + B cell lymphoma cells may be exploited to provide therapies that can slow or stop progression of the disease.
  • the therapy may reverse the course of the disease, in some cases even to the point of completely resolving—i.e., curing—the disease.
  • CD5 + B cell lymphoma cells having increased expression of one or more costimulatory molecules and/or other cell surface molecules may have diagnostic or investigative utility.
  • the first signal is the foreign antigen, which is presented by self-major histocompatibility complex (MHC) on the surface of an antigen presenting cell (APC).
  • MHC self-major histocompatibility complex
  • APC antigen presenting cell
  • TCR T cell receptor
  • costimulatory signals are antigen-independent and are provided to the naive T cell by the APC, through specific receptor-ligand interactions that promote, for example, T cell survival, clonal expansion, cytokine secretion, and effector function.
  • lymphocytes may fail to respond effectively to antigen stimulation. Such unresponsiveness of the adaptive immune system can result in immunologic tolerance.
  • costimulatory molecule refers to a member of a subset of cell surface molecules whose expression is necessary, in addition to presentation of a MHC I molecule, to generate a productive immune response, but whose expression is not independently sufficient to generate a productive immune response.
  • costimulatory molecules include, but are not limited to, members of the B7 family (including, for example, B7-1 (CD80), B7-2 (CD86), ICOS-L (B7RP1), and PDL-1), other molecules of the Ig superfamily (including, for example, CD2 and OX2), molecules of the TNF:TNFR subfamily that lack death domains (including, for example, CD40, OX40, CD27, 4-1BB, and CD30), and some integrins (including, for example, VLA-4, ICAM-1, and ICAM-3).
  • the increased expression of one or more cell surface molecules can be determined using any of many known methods, including any of the methods described herein.
  • methods include, but are not limited to, flow cytometry, immunohistochemistry, reverse transcriptase polymerase chain reaction (RT-PCR), including quantitative RT-PCR, and Northern blot analysis.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • CD5 + B cell lymphoma cells may be stimulated to increase expression of one or more of CD20, CD22, CD23, CD25, CD38, CD40, CD54, CD80, CD83, or CD86.
  • the CD5 + B cell lymphoma cells may be stimulated to increase expression of a cell surface molecule that is a target of a therapeutic agent such as, for example, a monoclonal antibody that specifically binds to the cell surface molecule.
  • a therapeutic agent such as, for example, a monoclonal antibody that specifically binds to the cell surface molecule.
  • rituximab is a monoclonal antibody that targets CD20 and has been shown to be an effective treatment for non-Hodgkin's lymphoma.
  • Rituximab binds to B cells that express CD20, thereby marking the rituximab-labeled cells for elimination by the immune system.
  • a treatment that includes, for example, (a) increasing expression of CD20 on CD5 + B cell lymphoma cells, and then (b) administering rituximab may permit rituximab to bind to CD5 + B cell lymphoma cells, thereby marking CD5 + B cell lymphoma cells for elimination by the immune system, and thereby rendering rituximab an effective treatment for CD5 + B cell lymphomas.
  • Additional cell surface molecules whose expression may be increased in CD5 + B cell lymphoma cells, and that may serve as a target for a therapeutic agent include, for example, CD22 and CD23.
  • CD5 + B cell lymphoma cells may be stimulated to produce a cytokine by contacting the CD5 + B cell lymphoma cells with an IRM effective to produce the cytokine in an amount that is greater than that produced by CD5 + B cell lymphoma cells not contacted by the IRM.
  • the IRM compound may be an agonist of one or more TLRs such as, for example, a TLR7 agonist, a TLR8 agonist, or an agonist of both TLR7 and TLR8.
  • the cytokine produced can include, but is not limited to, IL-1 ⁇ , IL-6, IL-8, IL-10, IL-12, TNF- ⁇ , GM-CSF, and combinations thereof.
  • the CD5 + B cell lymphoma cells may be contacted with an IRM compound in vitro, for example, in cell culture.
  • CD5 + B cell lymphoma cells may be contacted with an IRM compound in vivo—i.e., the CD5 + B cell lymphoma cells and IRM compound may be contacted in an organ, a tissue, or the blood.
  • the CD5 + B cell lymphoma cells may be contacted with an IRM compound in a subject by, for example, administering an IRM compound to a subject diagnosed as having a CD5 + B cell lymphoma.
  • the IRM directly to the subject allows the CD5 + B cell lymphoma cells, after being contacted with IRM, to activate autologous T cells—i.e., the subject's own T cells—thereby generating a T cell-dependent immune response against the CD5 + B cell lymphoma cells.
  • autologous T cells i.e., the subject's own T cells
  • a T cell-dependent immune response against the CD5 + B cell lymphoma cells By exploiting the subject's own T cell population to generate an immunological response to the CD5 + B cell lymphoma cells, one may be able to reduce or even eliminate certain risks associated with therapies that involve administering heterologous biological material (e.g., inflammation, rejection, etc.).
  • the IRM compounds may be administered via any suitable means, including, for example, parenterally, transdermally, intranasally, and orally. Suitable formulations for delivery of IRM compounds are described in detail below.
  • an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of CD5 + B cell lymphoma cells can be administered to a subject suffering from a CD5 + B cell lymphoma in a clinically effective amount.
  • a “clinically effective amount” is an amount effective to demonstrate one or more indications of clinical improvement. Such indications of clinical improvement can include any of those measurements applied in medical practice or laboratory research. See, for example, Cheson et al., National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood. 1996;87:4990-4997.
  • a clinically effective amount may be an amount effective to obtain a partial response (PR).
  • a “partial response” is at least about a 50% decrease in peripheral blood lymphocytes, lymphadenopathy, and/or splenomegaly, for at least two months.
  • a clinically effective amount may be an amount effective to obtain a complete response (CR).
  • a “complete response” is the absence of detectable leukemia or lymphoma cells.
  • a clinically effective amount may be an amount effective to prevent progressive disease (PD).
  • PD progressive disease
  • progressive disease is at least about a 50% increase in circulating lymphocytes or the progression to a more aggressive histology, as determined by known pathological criteria.
  • a clinically effective amount may be an amount effective to increase the likelihood or extent of long-term survival.
  • a clinically effective amount may be an amount that reduces or ameliorates at least one symptom or clinical sign associated with a CD5 + B cell lymphoma.
  • a clinically effective amount may be an amount sufficient to reduce the severity, extent, or number of cutaneous lymphoma deposits.
  • the effect of contacting one or more IRMs with CD5 + B cell lymphoma cells may be enhanced by further contacting the CD5 + B cell lymphoma cells with one or more additional immunomodulatory agents.
  • the IRM and one or more additional immunomodulatory agents may be considered a combination such as, for example, a therapeutic combination.
  • Components of such a combination may be said to be delivered “in combination” with one another if the components are provided in any manner that permits the biological effect of contacting one component with CD5 + B cell lymphoma cells to be sustained at least until another component is contacted with the CD5 + B cell lymphoma cells.
  • components may be delivered in combination with one another even if they are provided in separate formulations, delivered via different routes of administration, and/or administered at different times.
  • a suitable immunomodulatory agent may include, for example, interleukin-2 (“IL-2”).
  • IL-2 is a growth factor for antigen-stimulated T lymphocytes and is responsible for T cell clonal expansion after antigen recognition.
  • IL-2 can be obtained from any of many well-known sources.
  • clinical grade IL-2 can be commercially purchased, for example, from Chiron Corporation, San Francisco, Calif.
  • the CD5 + B cell lymphoma cells may be contacted with IL-2 in vitro, for example, in cell culture.
  • CD5 + B cell lymphoma cells may be contacted with IL-2 in vivo—i.e., the CD5 + B cell lymphoma cells and IL-2 may be contacted in an organ, a tissue, or the blood.
  • the CD5 + B cell lymphoma cells may be contacted with IL-2 in a subject by, for example, administering IL-2 to a subject suffering from a CD5 + B cell lymphoma.
  • the IL-2 may be administered via any suitable means, including, for example, parenterally, transdermally, intranasally, and orally. Suitable formulations for delivery of IL-2 are described below.
  • IL-2 used in any one embodiment will vary according to factors known in the art, including but not limited to, the physical and chemical nature of the IL-2, the physical and chemical nature of the IRM or IRMs provided in combination with the IL-2, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IL-2, whether any additional immunomodulatory agents are being administered in combination with the IL-2, and the species to which the formulation is being administered. Accordingly it is not practical to set forth generally the amount that constitutes an amount of IL-2 effective for all possible applications. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • IL-2 may be administered to a subject following procedures similar to those outlined by Rosenberg et al. on the administration of IL-2 for the treatment of melanoma and renal cell carcinoma. Rosenberg et al., JAMA, 1994;271:907-913.
  • Another suitable immunomodulatory agent may include, for example, a protein kinase C (PKC) agonist.
  • PKC protein kinase C
  • PKC agonists include, but are not limited to, phorbol esters (Totterman et al., Nature, 1980;288: 176-178) and bryostatin-1 (Drexler et al., Blood, 1989;74: 1747-1757).
  • Physiological ligands of molecules on the surface of lymphoma cells also can serve as PKC agonists by inducing signal transduction through the cell surface molecules, resulting in the activation of members of the PKC family of proteins.
  • antibodies against certain molecules on the surface of lymphoma cells can also serve as PKC agonists.
  • Such antibodies include, for example, antibodies against MHC class I molecules and antibodies to surface Ig.
  • the CD5 + B cell lymphoma cells may be contacted with a PKC agonist in vitro, for example, in cell culture.
  • CD5 + B cell lymphoma cells may be contacted with a PKC agonist in vivo—i.e., the CD5 + B cell lymphoma cells and a PKC agonist may be contacted in an organ, a tissue, or the blood.
  • the CD5 + B cell lymphoma cells may be contacted with a PKC agonist in a subject by, for example, administering a PKC agonist to a subject suffering from a CD5 + B cell lymphoma.
  • the PKC agonist may be administered via any suitable means, including, for example, parenterally, transdermally, intranasally, and orally. Suitable formulations for delivery of a PKC agonist are described below.
  • a PKC agonist used in any one embodiment will vary according to factors known in the art including but not limited to the physical and chemical nature of the PKC agonist, the physical and chemical nature of the IRM or IRMs provided in combination with the PKC agonist, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the PKC agonist, whether any additional immunomodulatory agents are being administered in combination with the PKC agonist, and the species to which the formulation is being administered. Accordingly it is not practical to set forth generally the amount that constitutes an amount of PKC agonist effective for all possible applications. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • CD5 + B cell lymphoma cells may be contacted with (a) one or more IRMs effective to increase the expression of at least one costimulatory molecule, (b) IL-2, and (c) a PKC agonist.
  • IRMs effective to increase the expression of at least one costimulatory molecule
  • IL-2 costimulatory molecule
  • PKC agonist a PKC agonist
  • each component of the combination may be provided in a single formulation that includes all of the components.
  • the combination may be provided in two or more formulations, each of which may contain a component of the combination alone or together with one or both of the other components. If the combination is provided in a plurality of formulations, the various formulations may be of similar or dissimilar composition.
  • each formulation may be of similar or dissimilar form (e.g., aerosol, gel, cream, solution, etc.) and may be administered via similar or dissimilar delivery routes (e.g., injection, transdermal, intravenous, etc).
  • delivery routes e.g., injection, transdermal, intravenous, etc.
  • the various components may be contacted with the CD5 + B cell lymphoma cells in any order.
  • a result of increasing the expression of at least one costimulatory molecule on the cell surface of CD5 + B cell lymphoma cells may include increasing proliferation—i.e., expansion—of CD5 + B cell lymphoma cell-specific (hereinafter, “lymphoma cell-specific”) cytotoxic T cells (“CTLs”).
  • Proliferation of lymphoma cell-specific CTLs may result from contacting lymphoma cell-specific CD8 + T cells with CD5 + B cell lymphoma cells having increased surface expression of costimulatory molecules.
  • Expression of costimulatory molecules on the surface of CD5+ B cell lymphoma cells may be increased by any suitable method including, for example, one or more of the methods described above.
  • the CD8 + T cells are CD5 + B cell lymphoma cell-specific—i.e., CD8 + T cells to which, or descendants of a CD8 + T cell to which, a CD5 + B cell lymphoma cell-specific antigen has previously been presented.
  • the CD8 + T cells are naive—i.e., CD8 + T cells to which, or descendants of a CD8 + T cell to which, no antigen (CD5 + B cell lymphoma-specific or otherwise) has been presented previously.
  • Lymphoma cell-specific CTLs activated by contact with CD5 + B cell lymphoma cells having increased expression of at least one costimulatory molecule may exhibit, for example, greater proliferation than that demonstrated by lymphoma cell-specific CTLs activated by contact with CD5 + B cell lymphoma cells that do not exhibit increased expression of a one or more costimulatory molecules.
  • the present invention also provides vaccines, methods of making vaccines, and methods of treating a subject by administering a vaccine.
  • vaccines include isolated CD5 + B cell lymphoma cells, or immunologically active portions thereof, in which the isolated CD5 + B cell lymphoma cells have been contacted with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface.
  • Isolated CD5 + B cell lymphoma cells also may be contacted with IL-2, a PKC agonist, or a combination of both IL-2 and a PKC agonist.
  • An immunologically active portion of a CD5 + B cell lymphoma cell can include, but is not limited to, a cell membrane preparation and/or a protein preparation from the isolated CD5 + B cell lymphoma cells.
  • a membrane preparation may include portions of the cell membrane from CD5 + B cell lymphoma cells and, for example, proteins embedded therein.
  • Vaccines may be made following any of the various procedures for the preparation of cell-based immunizations. For example, methods similar to those used for the preparation of cell-based vaccines against melanoma (see, for example, Wu et al., J Interferon Cytokine Res.
  • renal cancer cells see, for example, Vieweg et al., Urol. Clin. North Am. 2003 August; 30(3):633-43) or brain tumors (see, for example, Fecci et al., J. Neurooncol. 2003 August-September; 64(1-2): 161-76) may be used.
  • IRMs include compounds that possess potent immunomodulating activity including but not limited to antiviral and antitumor activity.
  • Certain IRMs modulate the production and secretion of cytokines.
  • certain IRM compounds induce the production and secretion of cytokines such as, e.g., Type I interferons, TNF- ⁇ , IL-1, IL-6, IL-8, IL-10, IL-12, MIP-1, and/or MCP-1.
  • certain IRM compounds can inhibit production and secretion of certain TH2 cytokines, such as IL-4 and IL-5. Additionally, some IRM compounds are said to suppress IL-1 and TNF (U.S. Pat. No. 6,518,265).
  • IRMs are small organic molecules (e.g., molecular weight under about 1000 Daltons, preferably under about 500 Daltons, as opposed to large biological molecules such as proteins, peptides, and the like) such as those disclosed in, for example, U.S. Pat. Nos.
  • IRMs include certain purine derivatives (such as those described in U.S. Pat. Nos. 6,376,501, and 6,028,076), certain imidazoquinoline amide derivatives (such as those described in U.S. Pat. No. 6,069,149), certain imidazopyridine derivatives (such as those described in U.S. Pat. No. 6,518,265), certain benzimidazole derivatives (such as those described in U.S. Pat. No. 6,387,938), certain derivatives of a 4-aminopyrimidine fused to a five membered nitrogen containing heterocyclic ring (such as adenine derivatives described in U.S. Pat. Nos.
  • IRMs include large biological molecules such as oligonucleotide sequences.
  • Some IRM oligonucleotide sequences contain cytosine-guanine dinucleotides (CpG) and are described, for example, in U.S. Pat. Nos. 6,194,388; 6,207,646; 6,239,116; 6,339,068; and 6,406,705.
  • CpG-containing oligonucleotides can include synthetic immunomodulatory structural motifs such as those described, for example, in U.S. Pat. Nos. 6,426,334 and 6,476,000.
  • Other IRM nucleotide sequences lack CpG sequences and are described, for example, in International Patent Publication No. WO 00/75304.
  • IRMs include biological molecules such as aminoalkyl glucosaminide phosphates (AGPs) and are described, for example, in U.S. Pat. Nos. 6,113,918; 6,303,347; 6,525,028; and 6,649,172.
  • AGPs aminoalkyl glucosaminide phosphates
  • any suitable IRM compound may be used to practice the invention.
  • reference to a compound can include the compound in any pharmaceutically acceptable form, including any isomer (e.g., diastereomer or enantiomer), salt, solvate, polymorph, and the like.
  • reference to the compound can include each of the compound's enantiomers as well as racemic mixtures of the enantiomers.
  • a suitable IRM compound may be, for example, a small molecule IRM compound such as one of those described above.
  • Suitable small molecule IRM compounds include those having a 2-aminopyridine fused to a five membered nitrogen-containing heterocyclic ring such as, for example, imidazoquinoline amines including but not limited to amide substituted imidazoquinoline amines, sulfonamide substituted imidazoquinoline amines, urea substituted imidazoquinoline amines, aryl ether substituted imidazoquinoline amines, heterocyclic ether substituted imidazoquinoline amines, amido ether substituted imidazoquinoline amines, sulfonamido ether substituted imidazoquinoline amines, urea substituted imidazoquinoline ethers, thioether substituted imidazoquinoline amines, and 6-, 7-, 8-, or 9-aryl or heteroaryl substituted imidazoquinoline amines
  • the IRM compound can be a tetrahydroimidazoquinoline amine such as, for example, 4-amino-2-(ethoxymethyl)- ⁇ , ⁇ -dimethyl-6,7,8,9-tetrahydro-1H-imidazo[4,5-c]quinoline-1-ethanol.
  • the IRM compound may be an imidazoquinoline amine.
  • the IRM may be 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin4-amine.
  • the IRM compound may be a sulfonamide substituted imidazoquinoline amine such as, for example, N-[4-(4-amino-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)butyl]methanesulfonamide.
  • a sulfonamide substituted imidazoquinoline amine such as, for example, N-[4-(4-amino-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)butyl]methanesulfonamide.
  • IRMs Various combinations of IRMs can be used if desired.
  • the IRM compound may be provided in any formulation suitable for administration to a subject. Suitable types of formulations are described, for example, in U.S. Pat. No. 5,736,553; U.S. Pat. No. 5,238,944; U.S. Pat. No. 5,939,090; U.S. Pat. No. 6,365,166; U.S. Pat. No. 6,245,776; U.S. Pat. No. 6,486,186; European Patent No. EP 0 394 026; and U.S. Patent Publication No. 2003/0199538.
  • the compound may be provided in any suitable form including but not limited to a solution, a suspension, an emulsion, or any form of mixture.
  • the compound may be delivered in formulation with any pharmaceutically acceptable excipient, carrier, or vehicle.
  • the compound may be provided in a formulation suitable for topical administration. Suitable types of formulations for topical delivery of, for example, certain IRM compounds are described, e.g., in International Patent Publication No. WO 03/045391.
  • the formulation may be delivered in any conventional dosage form including but not limited to a cream, an ointment, an aerosol formulation, a non-aerosol spray, a gel, a lotion, a tablet, a lozenge, an elixir, and the like.
  • the formulation may further include one or more additives including but not limited to adjuvants, skin penetration enhancers, colorants, fragrances, moisturizers, thickeners, and the like.
  • composition of a formulation suitable for practicing the invention will vary according to factors known in the art including but not limited to the physical and chemical nature of the IRM compound, the nature of the carrier, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IRM compound, whether the IRM is being administered in combination with one or more additional agents, and the species to which the formulation is being administered. Accordingly, it is not practical to set forth generally the composition of a formulation effective for all possible applications and all possible embodiments of the invention. Those of ordinary skill in the art, however, can readily determine an appropriate formulation with due consideration of such factors.
  • the methods of the present invention include administering IRM to a subject in a formulation of, for example, from about 0.0001% to about 10% (unless otherwise indicated, all percentages provided herein are weight/weight with respect to the total formulation) to the subject, although in some embodiments the IRM compound may be administered using a formulation that provides IRM compound in a concentration outside of this range.
  • the method includes administering to a subject a formulation that includes from about 0.01% to about 5% IRM compound, for example, a formulation that includes about 5% IRM compound.
  • an amount of an IRM compound effective for practicing the invention will vary according to factors known in the art including but not limited to the physical and chemical nature of the IRM compound, the nature of the carrier, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IRM compound, whether the IRM is being administered in combination with one or more additional agents, and the species to which the formulation is being administered. Accordingly, it is not practical to set forth generally the amount that constitutes an amount of IRM compound effective for all possible applications and all possible embodiments of the invention. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • the methods of the present invention include administering sufficient IRM compound to provide a dose of, for example, from about 100 ng/kg to about 50 mg/kg to the subject, although in some embodiments the methods may be performed by administering IRM compound in a dose outside this range.
  • the method includes administering sufficient IRM compound to provide a dose of from about 10 ⁇ g/kg to about 5 mg/kg to the subject, for example, a dose of from about 100 ⁇ g/kg to about 1 mg/kg.
  • the dosing regimen may depend at least in part on many factors known in the art including but not limited to the physical and chemical nature of the IRM compound, the nature of the carrier, the amount of IRM being administered, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IRM compound, whether the IRM is being administered in combination with one or more additional agents, and the species to which the formulation is being administered. Accordingly it is not practical to set forth generally the dosing regimen effective for all possible applications and all possible embodiments of the invention. Those of ordinary skill in the art, however, can readily determine an appropriate dosing regimen with due consideration of such factors.
  • the IRM compound may be administered, for example, from a single dose to multiple doses per day. In certain embodiments, the IRM compound may be administered from about three times per week to about once per day. In one particular embodiment, the IRM compound is administered once per day.
  • Antibodies Phycoerythrin- or FITC-labeled CD80 (B7-1), CD86 (B7-2), CD54 (ICAM-1), CD83, 4-1BB ligand (4-1BBL), CD5, and CD19 antibodies were purchased from BD Pharmingen (San Francisco, Calif.). Phycoerythrin-labeled ICOS-L and PDL-1 (B7-H1) antibodies were obtained from eBioscience (San Diego, Calif.).
  • CLL and T cells were isolated from fresh blood by negative selection (RosetteSep, StemCell Technologies, Vancouver, BC) as described by Gitelson et al. (Gitelson et al., Clin. Cancer Res. 2003;99:1656-1665).
  • CLL cells Purified CLL cells (1.5 ⁇ 10 6 cells/mL) were cultured in serum-free AIM-V medium plus 2-mercaptoethanol (2-ME, 5 ⁇ 10 ⁇ 5 M) (Sigma Chemical Co.) in 6- or 24-well plates (Becton-Dickinson Labware, Franklin Lake, N.J.) for 3-4 days at 37° C. in 5% CO 2 .
  • CLL cells were activated by adding the Negative Control compound (1 ⁇ g/mL), IRM1 (1 ⁇ g/mL), IL-2 (5000 U/mL), PDB (100 ng/mL), or bryostatin (20 nmol), as appropriate.
  • the Negative Control compound did not have measurable effects on CLL cells and, consequently, AIM-V medium, alone, was used as a control for the majority of the experiments.
  • MLRs Mixed Lymphocyte Responses
  • Cytokine levels in culture supernatants were determined by a multi-analysis fluorescent bead assay system available from Luminex Corp., Austin, Tex., under the tradename LUMINEX-100 SYTEM.
  • a 5-plex human cytokine kit for IFN- ⁇ , IL-2, IL-4, IL-10 and TNF- ⁇ measurement was used, according to the manufacturer's instructions (R&D Systems, Minneapolis, Minn.).
  • Individual cytokine concentrations were determined from standard curves using software available from BioRad, Mississauga, Ontario, under the tradename BIO-PLEX 2.0. The assay was linear between 30 and 1000 ⁇ g/mL for each cytokine.
  • CLL cells from the indicated number of patients were cultured in IRM1 (1 ⁇ g/mL) for 3-4 days, and then assayed for expression of the costimulatory molecules indicated on the x-axis (at an intensity greater than the first decade of log fluorescence) of FIG. 1A .
  • the percentage of cells that expressed each costimulatory molecule and the mean fluorescence intensity (MFI) of expression were measured by flow cytometry. The “fold-increase” was then calculated from the ratio of these measurements to the percentage and MFI of control cells cultured without activating agents. The average and standard error of these relative increases in costimulatory molecule expression are shown in the FIG. 1A .
  • CLL cells from the indicated number of patients were cultured in IRM1, LPS (100 ⁇ g/mL), poly (I:C) (100 ⁇ g/mL) and IFN- ⁇ 2B (500 U/mL) for 48 hours. Relative increases in the expression of CD80 and CD86 were calculated as described for FIG. 1A , and are shown in FIG. 1B .
  • TLR2 and TLR4 are activated by bacterial LPSs while TLR3 is activated by viral double-stranded RNA and poly (I:C) (Gordon, Cell. 2002;111:927-930).
  • I:C viral double-stranded RNA and poly (I:C)
  • FIG. 1B LPS or poly (I:C) rarely affected costimulatory molecule expression by CLL cells ( FIG. 1B ).
  • IRM1 is one of a class of IRMs known to stimulate the release of IFN- ⁇ from DCs or monocytes (Gibson et al., Cell Immunol. 2002;218:74-86), the effects of IRM1 were unlikely to be mediated indirectly by this cytokine since costimulatory molecule expression by CLL cells did not change significantly after direct stimulation with IFN- ⁇ 2B ( FIG. 1B ).
  • FIG. 2A shows a characteristic example. The numbers in the dot plots in the upper and lower rows are the percentages of CD80 + and CD86 + CLL cells, respectively.
  • FIG. 2B is a graphical representation of the percentage of CLL cells expressing the different costimulatory molecules (determined by the percentage of cells with staining intensity above the first decade of log fluorescence) from the number of patients indicated in the graph legend. The average and standard error for each of these measurements are shown in the graphs.
  • FIG. 2C is a graphical representation of the mean fluorescence index (MFI) of expression of the different costimulatory molecules determined for CLL cells from the number of patients indicated in the graph legend. The average and standard error for each of these measurements are shown in the graphs. Only the MFI of CD54 expression (divided by 10) is shown since essentially all CLL cells express this molecule.
  • the numbers over the double-headed arrows represent the p-values for the differences between sample means.
  • IL-2 and IRM1 both increased the percentage of CLL cells that expressed CD80 and CD86, as well as the mean fluorescence intensity (MFI) of expression of these molecules ( FIG. 2 ).
  • MFI mean fluorescence intensity
  • FIG. 2A The effects of IL-2 and IRM1 on costimulatory molecule expression were additive ( FIG. 2A , right dot-plots and FIG. 2B ,C), suggesting they were mediated by different mechanisms.
  • the MFI for the costimulatory molecules CD80, CD86, CD54, and CD83 shown in FIG. 2C indicate that IRM1 increased the expression of all four of these costimulatory molecules on CLL cells.
  • the magnitude of CD80 expression was especially increased by IRM 1 in combination with IL-2.
  • CLL cells were purified from individual patients and cultured alone or with IL-2, IRM1, IL-2 and IRM1, PDB, PDB and IL-2, PDB and IRM1, or PDB, IL-2, and IRM1. After 3-4 days, these cells were harvested, washed extensively, irradiated (2500 cGy) and used to stimulate allogenic or autologous T cells from CLL patients (obtained at the same time as the CLL cells and rested in culture until added to the Mixed Lymphocytic Response (MLR) assay) after 5-6 days of culture, alamar blue was added and proliferation was measured in an optical density colorimetric microplate reader at wavelengths of 540 (reduced state) and 595 (oxidized state).
  • MLR Mixed Lymphocytic Response
  • results are shown in FIG. 3A .
  • the results from the T cell source that exhibited the greatest stimulation (after subtraction of the proliferation induced by non-activated CLL cell stimulators) from each individual experiment were used to generate the average proliferation and standard error from the number of patients indicated on the x-axis.
  • FIG. 3B is a graphical representation of the correlation of CD83 expression with T cell stimulatory ability.
  • CLL cells were treated with IL-2 and IRM1 for 4-5 days. The percentage of cells expressing CD83 and the MFI of CD83 expression were then determined by flow cytometry. The activated CLL cells were then irradiated and used to stimulate autologous or allogeneic T cells in MLRs. The initial percentages of CD83 + CLL cells (left panel) and the MFI of CD83 expression (right panel) from 19 different patients were then correlated with the measured proliferation in the MLRs. The best straight line has intercept 10.692 and slope 0.0598; the associated P-value is 0.0153.
  • FIG. 4A shows CLL cells from a representative patient that were cultured alone (left panels) or with IRM1, IL-2, and PDB (right panels) for 3 days.
  • CD80, CD83, CD54, and CD86 expression were then determined by flow cytometry.
  • the percentages in the dot-plots refer to CD80 (sum of the right and left upper quadrants) (top panels) and to CD86 (sum of the right upper and lower quadrants) (bottom panels).
  • FIG. 4B is a summary of the results of flow cytometric evaluation of the percentage of CLL cells expressing CD80, CD83, CD54, and CD86 (and the MFI of expression) after culture alone, with PDB, PDB and IL-2, PDB and IRM1, or PDB, IL-2 and IRM1 from the indicated number of patients in the graph legend. The average and standard error are shown. Only the MFI of CD54 expression is shown since essentially all CLL cells express this molecule.
  • FIG. 4C is a summary of similar flow cytometric evaluation of ICOS-L, 4-1BBL, and PDL-1 expression. The numbers over the double-headed arrows are the p-values for the differences between sample means.
  • results shown in FIG. 4 indicated that the combination of PDB and IRM1 caused nearly 100% of CLL cells to acquire CD80, CD86, and CD83 expression.
  • the addition of IL-2 affected mainly the magnitude of CD80 and CD54 expression.
  • PDB, with or without IL-2, and/or IRM1 increased the expression of 4-1BBL and PDL-1 but not to the same extent as CD80, CD86, CD54, and CD83.
  • CLL cells activated with PDB, IL-2, and IRM1 were reflected in the ability of these cells to stimulate T cell proliferation ( FIG. 3A ).
  • CLL cells stimulated with PDB (without IL-2) were weak stimulators of T cell proliferation ( FIG. 3A ).
  • CLL cells and T cells were isolated from a CLL patient, suspended at concentrations of 10 6 cells/mL and mixed in a 1:1 ratio.
  • the cell mixtures were cultured alone, or in the presence of IL-2, IL-2 and IRM1, bryostatin, bryostatin and IL-2, bryostatin and IRM1, or bryostatin, IL-2, and IRM1.
  • FIG. 5A after 5 days, the percentages of CD5 + CD19 + tumor cells (indicated by the numbers in the right upper quadrants of the dot-plots) and CD5 + CD19 ⁇ T cells were determined by flow cytometry.
  • FIG. 5B these percentages and the total numbers of viable cells (determined by manual counting in a hemocytometer) were used to calculate the remaining absolute numbers of CLL cells in the cultures.
  • IRM1 whether alone or in combination with IL-2 and/or bryostatin, induced autologous T cells to kill CLL cells in vitro.
  • the combination of IRM1, IL-2, and bryostatin enabled autologous T cells to achieve 100% clearance of CLL cells in 5 days.
  • a 71-year old Caucasian male was diagnosed with Rai Stage 0 CLL on the basis of a persistent elevated count of circulating monoclonal CD19 + CD5 + IgM lo lymphocytes, determined by flow cytometry.
  • CD38 was expressed by 45% of circulating CLL cells.
  • the white blood cell counts at the beginning and end of treatment with IRM2 were 36 ⁇ 10 6 cells/mL and 45 ⁇ 10 6 cells/mL, respectively. Other systemic chemotherapy, steroids, or radiation had not been administered previously.
  • the patient reported having recurrent nodular, erythematous lesions on his hands and arms for approximately eight years.
  • the lesions were usually removed by treatment with liquid nitrogen.
  • he was diagnosed with CLL he had several such lesions over his upper back ( FIG. 8A ) and arms.
  • One lesion was biopsied and found to contain a diffuse atypical lymphoid dermal infiltrate consisting of many small, round lymphocytes, without epidermotropism.
  • the lymphoid filtrate had a predominant CD20 + phenotype.
  • Molecular analysis on paraffin embedded tissue demonstrated a monoclonal B cell population, consistent with B cell lymphoma.
  • a 5% cream of IRM2 was applied to the affected area three times per week. After eight weeks, the size of the treated lesion had not changed significantly, although an area of hypopigmentation had formed that was pronounced of a halo nevus around a regressing melanoma deposit.
  • IRM2 cream Administration of 5% IRM2 cream was increased to once per day. The lesion disappeared after six weeks of treatment ( FIG. 6B ) and had not recurred by three months after treatment ceased. Neither untreated lymphomatous lesions nor circulating white blood cell count changed significantly over the course of the treatment.
  • CLL cells were isolated from fresh blood by negative selection (RosetteSep, StemCell Technologies, Inc., Vancouver, BC) as described in Gitelson et al., Clin. Can. Res., 9:1656-1665 (2003). Purified CLL cells (1.5 ⁇ 10 6 cells/mL) were cultured in serum-free AIM-V medium (GibcoBRL) for three days. IRM1 and Negative Control compound were used at final concentrations of 1 ⁇ g/mL.
  • CLL cells from eight different patients were purified and cultured with either 0.001 ⁇ g/mL, 0.01 ⁇ g/mL, 0.1 ⁇ g/mL, or 1.0 ⁇ g/mL of IRM3 for three days.
  • CD80 and CD83 expression was determined by flow cytometry as described above. Increases in CD80 and CD83 expression were computed by subtracting CD80 and CD83 expression, respectively, from control CLL cultures to which no IRM3 was added. Results are presented in FIG. 8 .
  • CLL cells were collected from patients and cultured for 2-3 days with IRM3 (1 ⁇ g/mL) and without (control). The percentages of cells that expressed CD80, CD83, CD86, and CD38 were determined by flow cytometry. The MFI of expression of CD54 and CD25 was determined because virtually all CLL cells express these molecules. Changes in MFI of expression or the percentage of cells expressing a particular cell surface molecule were determined by subtracting values obtained from control cultures from the values obtained from cells cultured with IRM3. Results are presented in FIG. 9 .
  • CLL cells were purified and cultured either without (control) or with IRM3 (1 ⁇ g/mL) and IL-2 (5000 U/mL). After 48 hours, the MFI of CD20 expression was determined by flow cytometry. Results are presented in FIG. 10 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention provides methods for increasing expression of cell surface molecules of CD5+ B cell lymphoma cells by contacting cells with immune response modifiers. The invention also provides methods for the treatment of CD5+ B cell lymphomas, including chronic lymphocytic leukemia and small lymphocytic lymphoma, by administering immune response modifier compounds to a subject in need of such treatment. Suitable immune response modifier compounds include agonists of TLR7 and/or TLR8.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims priority to U.S. Provisional Patent Application Ser. No. 60/500,478, filed Sep. 5, 2003, and U.S. Provisional Patent Application Ser. No. 60/561,440, filed Apr. 12, 2004.
  • BACKGROUND
  • The peripheral B cell neoplasm chronic lymphoid leukemia/small lymphocytic lymphoma represents the most common lymphoid leukemia. As the name implies, presentation can be as either leukemia or lymphoma. However, the two presentations of this neoplasm, chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL), are morphologically, phenotypically, and genotypically indistinguishable, differing only in the degree of peripheral blood lymphocytosis.
  • CLL is the most common leukemia of adults in the Western world. In CLL, the peripheral blood contains small, round lymphocytes with scant cytoplasm. Involvement of the bone marrow is observed in all cases of CLL and most cases of SLL, taking the form of interstitial infiltrates or nonparatubular aggregates of small lymphocytes. The tumor cells in CLL and SLL express the pan B cell markers CD29 and CD20. In addition, CD5-a T cell marker that is expressed only on a small subset of normal B cells—is present on the tumor cells. The immunophenotype of CLL cells is unique. CLL cells co-express the B lymphocyte lineage marker CD19 and the T lymphocyte marker CD5. CLL cells also exhibit a characteristic level of expression of immunoglobulin receptor. Tumor cells typically also have low-level surface expression of Ig heavy chain, with either kappa or lambda light chains.
  • CLL is a clonal malignancy of B lymphocytes. The disease is usually indolent, with slowly progressive accumulation of long-lived small lymphocytes that are immunoincompetent and respond poorly to antigenic stimulation. CLL is incurable with conventional cytotoxic chemotherapy (Cheson et al., Blood 1996; 87:4990-4997; and Keating et al., Blood 1993; 81:2878-2884). The hallmark of CLL is isolated lymphocytosis. The white blood cell count is usually greater than 20,000 μL and may be markedly elevated to several hundred thousand. The diagnostic requirement for CLL is an absolute lymphocyte count of greater than 4000/mm3. CLL is manifested clinically by immunosuppression, bone marrow failure, and organ infiltration with lymphocytes. Immunodeficiency is also related to inadequate antibody production by the abnormal B cells. With advanced disease, CLL may cause damage by direct tissue infiltration.
  • In some cases, patients may develop cutaneous lymphoma deposits—erythomatous lesions on the skin. The lesions may contain an atypical lymphoid dermal infiltrate of small, round B cells.
  • SUMMARY
  • It has been found that certain small molecule immune response modifiers (IRMs) can be useful for increasing the expression of molecules on the surface of CD5+ B cell lymphoma cells. Thus, certain IRMs can be used for treating a CD5+ B cell lymphomas such as, for example, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), mantle cell lymphoma, or splenic lymphoma with villous lymphocytes.
  • Accordingly, the invention provides a method of treating a CD5+ B cell lymphoma. Generally, the method includes administering to a subject an IRM compound in an amount effective to ameliorate at least one symptom or clinical sign of the CD5+ B cell lymphoma. In some embodiments, administering the IRM compound may result in at least a 50% decrease in peripheral blood lymphocytes, lymphadenopathy, or splenomegaly for at least two months. In other embodiments, administering the IRM compound can inhibit or even prevent the development of progressive disease, wherein progressive disease is at least a 50% increase in circulating lymphocytes or a progression to a more aggressive histology. In still other embodiments, administering the IRM compound can resolve nodular, erythematous lesions associated with CD5+ B cell lymphoma.
  • In another aspect, the invention provides a method of increasing the expression of at least one cell surface molecule of CD5+ B cell lymphoma cells. Generally, the method includes contacting the CD5+ B cell lymphoma cells with at least one IRM effective for increasing the expression of at least one cell surface molecule of the CD5+ B cell lymphoma cells. In some embodiments, the cell surface molecule may be a costimulatory molecule.
  • In another aspect, the present invention also provides a method of stimulating CD5+ B cell lymphoma cells to produce a cytokine by contacting the CD5+ B cell lymphoma cells with an IRM effective for inducing production of a cytokine above a level produced by the CD5+ B cell lymphoma cells not contacted by the IRM. In some embodiments, the cytokine may be IL-1β, IL-6, IL-8, IL-10, IL-12, TNF-α, GM-CSF, or combinations thereof.
  • In another aspect, the invention provides a method of increasing proliferation of CD5+ B cell lymphoma-specific cytotoxic T cells. Generally, the method includes contacting CD5+ B cell lymphoma cells with an IRM effective to increase the expression of at least one costimulatory molecule on the surface of CD5+ B cell lymphoma cells, and then contacting CD8+ T cells with the CD5+ B cell lymphoma cells, thereby activating the CD8+ T cells; wherein the activated T cells are CD5+ B cell lymphoma-specific cytotoxic T cells and demonstrate increased proliferation compared to T cells contacted with CD5+ B cell lymphoma cells that have not been contacted with an IRM.
  • In some embodiments, the CD8+ T cells are CD5+ B cell lymphoma cell-specific. In other embodiments, the CD8+ T cells are naive.
  • In some embodiments, the IRM compound may be administered to a subject diagnosed as having a CD5+ B cell lymphoma so that the activated CD5+ B cell lymphoma-specific cytotoxic T cells are autologous CD5+ B cell lymphoma-specific cytotoxic T cells.
  • In some embodiments, the CD5+ B cell lymphoma cells may be further contacted with one or more additional immunomodulating agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • In another aspect, the present invention also provides a method of increasing the killing of CD5+ B cell lymphoma cells by cytotoxic T cells. Generally, the method includes contacting CD5+ B cell lymphoma cells with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of the CD5+ lymphoma cells, and then contacting CD8+ T cells with the CD5+ B cell lymphoma cells, thereby activating the CD8+ T cells; wherein the activated CD8+ T cells are CD5+ B cell lymphoma-specific cytotoxic T cells and demonstrate increased killing of CD5+ B cell lymphoma cells compared to T cells contacted with CD5+ B cell lymphoma cells that have not been contacted with an IRM.
  • In some embodiments, the CD8+ T cells are CD5+ B cell lymphoma cell-specific. In other embodiments, the CD8+ T cells are naive.
  • In some embodiments, the IRM compound may be administered to a subject diagnosed as having a CD5+ B cell lymphoma so that the activated CD5+ B cell lymphoma-specific cytotoxic T cells are autologous CD5+ B cell lymphoma-specific cytotoxic T cells.
  • In some embodiments, the CD5+ B cell lymphoma cells may be further contacted with one or more additional immunomodulating agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • In another aspect, the present invention also provides a method of treating a subject suffering from a CD5+ B cell lymphoma including administering to the subject an IRM effective to increase the expression of at least one cell surface molecule of the CD5+ B cell lymphoma cells.
  • In another aspect, the present invention also provides a vaccine that includes isolated CD5+ B cell lymphoma cells, or an immunologically active portion thereof, wherein the isolated CD5+ B cell lymphoma cells have been contacted with an IRM effective to increase the expression of at least one cell surface molecule of the CD5+ B cell lymphoma cells. In certain embodiments, the CD5+ B cell lymphoma cells may be further contacted with one or more additional immunomodulatory agents such as, for example, IL-2 and/or a protein kinase C agonist.
  • In another aspect, the present invention also provides a method of preparing a vaccine including contacting isolated CD5+ B cell lymphoma cells with an IRM effective to increase the expression of at least one molecule on the surface of the CD5+ B cell lymphoma cells. Some embodiments may include further contacting isolated CD5+ B cell lymphoma cells with one or more additional immunomodulatory agents such as, for example, IL-2 or a protein kinase C agonist. In other embodiments, the isolated CD5+ B cell lymphoma cells may be derived from a subject diagnosed as having CLL or SLL.
  • In some embodiments, the CD5+ B cell lymphoma cells may be chronic lymphocytic leukemia (CLL) cells, small lymphocytic lymphoma cells (SLL), mantle cell lymphoma cells, splenic lymphoma with villous lymphocytes, or combinations thereof.
  • In some embodiments, the IRM is a TLR7 agonist. In some embodiments, the IRM is a TLR8 agonist. In still other embodiments, the IRM compound may be an agonist of both TLR7 and TLR8.
  • In some embodiments, the IRM may be an imidazoquinoline amine, a tetrahydroimidazoquinoline amine, an imidazopyridine amine, a 1,2-bridged imidazoquinoline amine, a 6,7-fused cycloalkylimidazopyridine amine, an imidazonaphthyridine amine, a tetrahydroimidazonaphthyridine amine, an oxazoloquinoline amine, a thiazoloquinoline amine, an oxazolopyridine amine, a thiazolopyridine amine, an oxazolonaphthyridine amine, or a thiazolonaphthyridine amine. In certain embodiments, the IRM is an imidazoquinoline amine such as, for example, 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine. In alternative embodiments, the IRM is a tetrahydroimidazoquinoline amine such as, for example, 4-amino-2-(ethoxymethyl)-α,α-dimethyl-6,7,8,9-tetrahydro-1H-imidazo[4,5-c]quinoline-1-ethanol hydrate. In still other embodiments, the IRM is a sulfonamide substituted imidazoquinoline amine such as, for example, N-[4-(4-amino-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)butyl]methanesulfonamide.
  • In some embodiments, at least one cell surface molecule whose expression is increased may be CD20, CD22, or CD23, and the method further includes administering to the subject a therapeutic agent that has, as a target, the cell surface molecule whose expression is increased. In some embodiments, the expression of more than one cell surface molecule may be increased.
  • In some embodiments, at least one costimulatory molecule whose expression is increased may be CD40, CD54, CD80, CD83, CD86, CD25, or CD38. In some embodiments, the expression of more than one costimulatory molecule may be increased.
  • In some embodiments, the CD5+ B cell lymphoma cells may be contacted with an IRM in vitro. In othe r embodiments, the CD5+ B cell lymphoma cells may be contacted with an IRM in vivo such as, for example, in an organ, tissue, or blood of a subject.
  • Various other features and advantages of the present invention should become readily apparent with reference to the following detailed description, examples, claims and appended drawings. In several places throughout the specification, guidance is provided through lists of examples. In each instance, the recited list serves only as a representative group and should not be interpreted as an exclusive list.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1 a-b. Enhancement of costimulatory molecule expression on CLL cells by an IRM compound.
  • FIGS. 2A-C. Effect of an IRM compound (with or without IL-2) on costimulatory molecule expression by CLL cells.
  • FIGS. 3A-B. Effect of an IRM compound on the ability of CLL cells to stimulate cytotoxic T cell proliferation.
  • FIGS. 4A-C. Effect of an IRM compound, IL-2, and PKC agonists on costimulatory molecule expression by CLL cells.
  • FIGS. 5A-B. Induction of T cell cytotoxicity against autologous CLL cells by PKC agonists, IL-2, and IRM.
  • FIGS. 6A-B. Photographs of a lymphomatous skin deposit before treatment (FIG. 6A) and after treatment (FIG. 6B) with an IRM.
  • FIG. 7. Effect of IRM1 on costimulatory molecule expression by CLL cells.
  • FIG. 8A-B. Effect of IRM3 on expression of CD80 and CD83 by CLL cells.
  • FIG. 9. IRM3-Mediated changes in cell surface molecule expression in CLL cells.
  • FIG. 10. IRM3+IL-2 increases expression of CD20 on CLL cells.
  • DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS OF THE INVENTION
  • The present invention provides methods for treating CD5+ B cell lymphomas such as, for example, chronic lymphocytic leukemia (CLL). While a number of clinical observations suggest that CD5+ B cell lymphoma cells may be subject to T cell mediated immune recognition (see, for example, Ribera et al., Blood Cells 1987; 12:471-483; Ziegler-Heitbrock et al., Blood 1989; 73:1426-1430; Wierda et al., Blood 2000; 96:2917-2924; Gitelson et al., Clin Cancer Res. 2003; 99:1656-1665; and Pavletic et al., Bone Marrow Transplant 2000; 25:717-722), the weak immunogenicity of CD5+ B cell lymphoma cells has limited development of immunologically based treatment methods and, therefore, contributes to disease progression.
  • The present invention demonstrates, for the first time, that contacting CD5+ B cell lymphoma cells with an immune response modifier (IRM) compound may be useful for treating CD5+ B cell lymphomas. IRM compounds appear to act through basic immune system mechanisms known as toll-like receptors (TLRs) to induce selected biosynthesis of certain cytokines, chemokines and costimulatory molecules. Thus, certain IRM compounds can selectively induce certain aspects—and/or inhibit other aspects—of the immune system. In particular, IRM compounds may increase the expression of cell surface molecules of CD5+ B cell lymphoma cells, enhance the immunogenicity of CD5+ B cell lymphoma cells, and provide a new immunotherapeutic approach for the treatment of CD5+ B cell lymphomas. In some cases, the cell surface molecule whose expression is increased may be a costimulatory molecule. Increasing the cell surface expression of costimulatory molecules may allow the CD5+ B cell lymphoma cells to become competent antigen presenting cells (APCs) capable of initiating and/or maintaining tumor-reactive T cell activity.
  • As used herein, the following terms shall have the following meanings:
  • “Ameliorate” refers to any reduction in the extent, severity, frequency, and/or likelihood of a symptom or clinical sign characteristic of a particular condition.
  • “CD5+ B cell lymphoma cells” refers to neoplastic cells having a unique immunophenotype that includes co-expression of CD19 and CD5. In addition to expressing the B lymphocyte lineage marker CD19, CD5+ B cell lymphoma cells also express the T lymphocyte marker CD5, which is typically expressed only on a small subset of normal B cells. CD5+ B cell lymphoma cells include, for example, chronic lymphocytic leukemia (CLL) cells, small lymphocytic lymphoma (SLL) cells, mantle cell lymphoma cells, and splenic lymphoma with villous lymphocytes. In some embodiments, the CD5+ B cell lymphoma cells are CLL cells or SLL cells. In certain specific embodiments, the CD5+ B cell lymphoma cells are CLL cells.
  • “Cell surface molecule” refers to a molecule that is expressed on the surface of a cell and may be used to determine the cell's lineage or otherwise may be used to distinguish one cell or cell type from another.
  • “Patient” or “Subject” includes, for example, animals such as, but not limited to, humans, non-human primates, rodents, dogs, cats, horses, pigs, sheep, goats, or cows.
  • “Sign” or “clinical sign” refers to an objective physical finding relating to a particular condition capable of being found by one other than the patient.
  • “Symptom” refers to any subjective evidence of disease or of a patient's condition.
  • Unless otherwise specified, “a,” “an,” “the,” and “at least one” are used interchangeably and mean one or more than one.
  • Certain IRM compounds such as, for example, agonists of TLR7 and/or TLR8 can increase the expression of a number of cell surface molecules (including, e.g., costimulatory molecules) of CD5+ B cell lymphoma cells, which can result in a more potent immune response being raised against the CD5+ B cell lymphoma cells. Thus, increasing expression of cell surface molecules of CD5+ B cell lymphoma cells may be exploited to provide therapies that can slow or stop progression of the disease. In certain embodiments, the therapy may reverse the course of the disease, in some cases even to the point of completely resolving—i.e., curing—the disease.
  • In addition to having therapeutic utility, CD5+ B cell lymphoma cells having increased expression of one or more costimulatory molecules and/or other cell surface molecules may have diagnostic or investigative utility.
  • Two signals are required for the induction of cell proliferation and cytokine production in naive T cells. The first signal is the foreign antigen, which is presented by self-major histocompatibility complex (MHC) on the surface of an antigen presenting cell (APC). The antigenic peptide-MHC complex interacts with a T cell receptor (TCR) on the surface of the naive T cell, thereby providing antigen specificity to the immune response. The second signal is a “costimulatory” signal. Costimulatory signals are antigen-independent and are provided to the naive T cell by the APC, through specific receptor-ligand interactions that promote, for example, T cell survival, clonal expansion, cytokine secretion, and effector function.
  • In the presence of both signals, a productive adaptive immune response may be generated. In the absence of the costimulatory signal, however, lymphocytes may fail to respond effectively to antigen stimulation. Such unresponsiveness of the adaptive immune system can result in immunologic tolerance.
  • Accordingly, as used herein, “costimulatory molecule” refers to a member of a subset of cell surface molecules whose expression is necessary, in addition to presentation of a MHC I molecule, to generate a productive immune response, but whose expression is not independently sufficient to generate a productive immune response. Examples of costimulatory molecules include, but are not limited to, members of the B7 family (including, for example, B7-1 (CD80), B7-2 (CD86), ICOS-L (B7RP1), and PDL-1), other molecules of the Ig superfamily (including, for example, CD2 and OX2), molecules of the TNF:TNFR subfamily that lack death domains (including, for example, CD40, OX40, CD27, 4-1BB, and CD30), and some integrins (including, for example, VLA-4, ICAM-1, and ICAM-3).
  • The increased expression of one or more cell surface molecules can be determined using any of many known methods, including any of the methods described herein. For example, such methods include, but are not limited to, flow cytometry, immunohistochemistry, reverse transcriptase polymerase chain reaction (RT-PCR), including quantitative RT-PCR, and Northern blot analysis.
  • In some embodiments, CD5+ B cell lymphoma cells may be stimulated to increase expression of one or more of CD20, CD22, CD23, CD25, CD38, CD40, CD54, CD80, CD83, or CD86.
  • In certain embodiments, the CD5+ B cell lymphoma cells may be stimulated to increase expression of a cell surface molecule that is a target of a therapeutic agent such as, for example, a monoclonal antibody that specifically binds to the cell surface molecule. In this way, the increase in cell surface molecule expression may be exploited in a treatment that is targeted against the cell surface molecule. For example, rituximab is a monoclonal antibody that targets CD20 and has been shown to be an effective treatment for non-Hodgkin's lymphoma. Rituximab binds to B cells that express CD20, thereby marking the rituximab-labeled cells for elimination by the immune system. Thus, a treatment that includes, for example, (a) increasing expression of CD20 on CD5+ B cell lymphoma cells, and then (b) administering rituximab may permit rituximab to bind to CD5+ B cell lymphoma cells, thereby marking CD5+ B cell lymphoma cells for elimination by the immune system, and thereby rendering rituximab an effective treatment for CD5+ B cell lymphomas. Additional cell surface molecules whose expression may be increased in CD5+ B cell lymphoma cells, and that may serve as a target for a therapeutic agent include, for example, CD22 and CD23.
  • In some embodiments, CD5+ B cell lymphoma cells may be stimulated to produce a cytokine by contacting the CD5+ B cell lymphoma cells with an IRM effective to produce the cytokine in an amount that is greater than that produced by CD5+ B cell lymphoma cells not contacted by the IRM. In some embodiments, the IRM compound may be an agonist of one or more TLRs such as, for example, a TLR7 agonist, a TLR8 agonist, or an agonist of both TLR7 and TLR8. The cytokine produced can include, but is not limited to, IL-1β, IL-6, IL-8, IL-10, IL-12, TNF-α, GM-CSF, and combinations thereof.
  • In some embodiments, the CD5+ B cell lymphoma cells may be contacted with an IRM compound in vitro, for example, in cell culture. In alternative embodiments, CD5+ B cell lymphoma cells may be contacted with an IRM compound in vivo—i.e., the CD5+ B cell lymphoma cells and IRM compound may be contacted in an organ, a tissue, or the blood. In such cases, the CD5+ B cell lymphoma cells may be contacted with an IRM compound in a subject by, for example, administering an IRM compound to a subject diagnosed as having a CD5+ B cell lymphoma. Administration of the IRM directly to the subject allows the CD5+ B cell lymphoma cells, after being contacted with IRM, to activate autologous T cells—i.e., the subject's own T cells—thereby generating a T cell-dependent immune response against the CD5+ B cell lymphoma cells. By exploiting the subject's own T cell population to generate an immunological response to the CD5+ B cell lymphoma cells, one may be able to reduce or even eliminate certain risks associated with therapies that involve administering heterologous biological material (e.g., inflammation, rejection, etc.).
  • The IRM compounds may be administered via any suitable means, including, for example, parenterally, transdermally, intranasally, and orally. Suitable formulations for delivery of IRM compounds are described in detail below.
  • In some embodiments, an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of CD5+ B cell lymphoma cells can be administered to a subject suffering from a CD5+ B cell lymphoma in a clinically effective amount. As used herein a “clinically effective amount” is an amount effective to demonstrate one or more indications of clinical improvement. Such indications of clinical improvement can include any of those measurements applied in medical practice or laboratory research. See, for example, Cheson et al., National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood. 1996;87:4990-4997. For example, a clinically effective amount may be an amount effective to obtain a partial response (PR). As used herein, a “partial response” is at least about a 50% decrease in peripheral blood lymphocytes, lymphadenopathy, and/or splenomegaly, for at least two months. A clinically effective amount may be an amount effective to obtain a complete response (CR). As used herein, a “complete response” is the absence of detectable leukemia or lymphoma cells. A clinically effective amount may be an amount effective to prevent progressive disease (PD). As used herein, “progressive disease” is at least about a 50% increase in circulating lymphocytes or the progression to a more aggressive histology, as determined by known pathological criteria. A clinically effective amount may be an amount effective to increase the likelihood or extent of long-term survival. Alternatively, a clinically effective amount may be an amount that reduces or ameliorates at least one symptom or clinical sign associated with a CD5+ B cell lymphoma. For example, a clinically effective amount may be an amount sufficient to reduce the severity, extent, or number of cutaneous lymphoma deposits.
  • In some embodiments, the effect of contacting one or more IRMs with CD5+ B cell lymphoma cells may be enhanced by further contacting the CD5+ B cell lymphoma cells with one or more additional immunomodulatory agents. In such embodiments, the IRM and one or more additional immunomodulatory agents may be considered a combination such as, for example, a therapeutic combination. Components of such a combination may be said to be delivered “in combination” with one another if the components are provided in any manner that permits the biological effect of contacting one component with CD5+ B cell lymphoma cells to be sustained at least until another component is contacted with the CD5+ B cell lymphoma cells. Thus, components may be delivered in combination with one another even if they are provided in separate formulations, delivered via different routes of administration, and/or administered at different times.
  • A suitable immunomodulatory agent may include, for example, interleukin-2 (“IL-2”). IL-2 is a growth factor for antigen-stimulated T lymphocytes and is responsible for T cell clonal expansion after antigen recognition. IL-2 can be obtained from any of many well-known sources. For example, clinical grade IL-2 can be commercially purchased, for example, from Chiron Corporation, San Francisco, Calif.
  • In some embodiments, the CD5+ B cell lymphoma cells may be contacted with IL-2 in vitro, for example, in cell culture. In alternative embodiments, CD5+ B cell lymphoma cells may be contacted with IL-2 in vivo—i.e., the CD5+ B cell lymphoma cells and IL-2 may be contacted in an organ, a tissue, or the blood. In such cases, the CD5+ B cell lymphoma cells may be contacted with IL-2 in a subject by, for example, administering IL-2 to a subject suffering from a CD5+ B cell lymphoma. The IL-2 may be administered via any suitable means, including, for example, parenterally, transdermally, intranasally, and orally. Suitable formulations for delivery of IL-2 are described below.
  • The precise amount of IL-2 used in any one embodiment will vary according to factors known in the art, including but not limited to, the physical and chemical nature of the IL-2, the physical and chemical nature of the IRM or IRMs provided in combination with the IL-2, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IL-2, whether any additional immunomodulatory agents are being administered in combination with the IL-2, and the species to which the formulation is being administered. Accordingly it is not practical to set forth generally the amount that constitutes an amount of IL-2 effective for all possible applications. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors. For example, IL-2 may be administered to a subject following procedures similar to those outlined by Rosenberg et al. on the administration of IL-2 for the treatment of melanoma and renal cell carcinoma. Rosenberg et al., JAMA, 1994;271:907-913.
  • Another suitable immunomodulatory agent may include, for example, a protein kinase C (PKC) agonist. Examples of PKC agonists include, but are not limited to, phorbol esters (Totterman et al., Nature, 1980;288: 176-178) and bryostatin-1 (Drexler et al., Blood, 1989;74: 1747-1757). Physiological ligands of molecules on the surface of lymphoma cells also can serve as PKC agonists by inducing signal transduction through the cell surface molecules, resulting in the activation of members of the PKC family of proteins. For example, antibodies against certain molecules on the surface of lymphoma cells can also serve as PKC agonists. Such antibodies include, for example, antibodies against MHC class I molecules and antibodies to surface Ig.
  • In some embodiments, the CD5+ B cell lymphoma cells may be contacted with a PKC agonist in vitro, for example, in cell culture. In alternative embodiments, CD5+ B cell lymphoma cells may be contacted with a PKC agonist in vivo—i.e., the CD5+ B cell lymphoma cells and a PKC agonist may be contacted in an organ, a tissue, or the blood. In such cases, the CD5+ B cell lymphoma cells may be contacted with a PKC agonist in a subject by, for example, administering a PKC agonist to a subject suffering from a CD5+ B cell lymphoma. The PKC agonist may be administered via any suitable means, including, for example, parenterally, transdermally, intranasally, and orally. Suitable formulations for delivery of a PKC agonist are described below.
  • The precise amount of a PKC agonist used in any one embodiment will vary according to factors known in the art including but not limited to the physical and chemical nature of the PKC agonist, the physical and chemical nature of the IRM or IRMs provided in combination with the PKC agonist, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the PKC agonist, whether any additional immunomodulatory agents are being administered in combination with the PKC agonist, and the species to which the formulation is being administered. Accordingly it is not practical to set forth generally the amount that constitutes an amount of PKC agonist effective for all possible applications. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • In certain embodiments, CD5+ B cell lymphoma cells may be contacted with (a) one or more IRMs effective to increase the expression of at least one costimulatory molecule, (b) IL-2, and (c) a PKC agonist. When CD5+ B cell lymphoma cells are contacted with the combination of such an IRM, IL-2, and a PKC agonist, each component of the combination may be provided in a single formulation that includes all of the components. Alternatively, the combination may be provided in two or more formulations, each of which may contain a component of the combination alone or together with one or both of the other components. If the combination is provided in a plurality of formulations, the various formulations may be of similar or dissimilar composition. Furthermore, each formulation may be of similar or dissimilar form (e.g., aerosol, gel, cream, solution, etc.) and may be administered via similar or dissimilar delivery routes (e.g., injection, transdermal, intravenous, etc). Also, if the components of the combination are provided in a plurality of formulations, the various components may be contacted with the CD5+ B cell lymphoma cells in any order.
  • In some embodiments, a result of increasing the expression of at least one costimulatory molecule on the cell surface of CD5+ B cell lymphoma cells may include increasing proliferation—i.e., expansion—of CD5+ B cell lymphoma cell-specific (hereinafter, “lymphoma cell-specific”) cytotoxic T cells (“CTLs”). Proliferation of lymphoma cell-specific CTLs may result from contacting lymphoma cell-specific CD8+ T cells with CD5+ B cell lymphoma cells having increased surface expression of costimulatory molecules. Expression of costimulatory molecules on the surface of CD5+ B cell lymphoma cells may be increased by any suitable method including, for example, one or more of the methods described above.
  • In some embodiments, the CD8+ T cells are CD5+ B cell lymphoma cell-specific—i.e., CD8+ T cells to which, or descendants of a CD8+ T cell to which, a CD5+ B cell lymphoma cell-specific antigen has previously been presented. In other embodiments, the CD8+ T cells are naive—i.e., CD8+ T cells to which, or descendants of a CD8+ T cell to which, no antigen (CD5+ B cell lymphoma-specific or otherwise) has been presented previously.
  • Lymphoma cell-specific CTLs activated by contact with CD5+ B cell lymphoma cells having increased expression of at least one costimulatory molecule may exhibit, for example, greater proliferation than that demonstrated by lymphoma cell-specific CTLs activated by contact with CD5+ B cell lymphoma cells that do not exhibit increased expression of a one or more costimulatory molecules.
  • In other aspects, the present invention also provides vaccines, methods of making vaccines, and methods of treating a subject by administering a vaccine. Such vaccines include isolated CD5+ B cell lymphoma cells, or immunologically active portions thereof, in which the isolated CD5+ B cell lymphoma cells have been contacted with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface. Isolated CD5+ B cell lymphoma cells also may be contacted with IL-2, a PKC agonist, or a combination of both IL-2 and a PKC agonist. An immunologically active portion of a CD5+ B cell lymphoma cell can include, but is not limited to, a cell membrane preparation and/or a protein preparation from the isolated CD5+ B cell lymphoma cells. Thus, for example, a membrane preparation may include portions of the cell membrane from CD5+ B cell lymphoma cells and, for example, proteins embedded therein. Vaccines may be made following any of the various procedures for the preparation of cell-based immunizations. For example, methods similar to those used for the preparation of cell-based vaccines against melanoma (see, for example, Wu et al., J Interferon Cytokine Res. 2001 December; 21(12): 1117-27), renal cancer cells (see, for example, Vieweg et al., Urol. Clin. North Am. 2003 August; 30(3):633-43) or brain tumors (see, for example, Fecci et al., J. Neurooncol. 2003 August-September; 64(1-2): 161-76) may be used.
  • IRMs include compounds that possess potent immunomodulating activity including but not limited to antiviral and antitumor activity. Certain IRMs modulate the production and secretion of cytokines. For example, certain IRM compounds induce the production and secretion of cytokines such as, e.g., Type I interferons, TNF-α, IL-1, IL-6, IL-8, IL-10, IL-12, MIP-1, and/or MCP-1. As another example, certain IRM compounds can inhibit production and secretion of certain TH2 cytokines, such as IL-4 and IL-5. Additionally, some IRM compounds are said to suppress IL-1 and TNF (U.S. Pat. No. 6,518,265).
  • Certain IRMs are small organic molecules (e.g., molecular weight under about 1000 Daltons, preferably under about 500 Daltons, as opposed to large biological molecules such as proteins, peptides, and the like) such as those disclosed in, for example, U.S. Pat. Nos. 4,689,338; 4,929,624; 4,988,815; 5,037,986; 5,175,296; 5,238,944; 5,266,575; 5,268,376; 5,346,905; 5,352,784; 5,367,076; 5,389,640; 5,395,937; 5,446,153; 5,482,936; 5,693,811; 5,741,908; 5,756,747; 5,939,090; 6,039,969; 6,083,505; 6,110,929; 6,194,425; 6,245,776; 6,331,539; 6,376,669; 6,451,810; 6,525,064; 6,541,485; 6,545,016; 6,545,017; 6,558,951; 6,573,273; 6,656,938; 6,660,735; 6,660,747; 6,664,260; 6,664,264; 6,664,265; 6,667,312; 6,670,372; 6,677,347; 6,677,348; 6,677,349; 6,683,088; 6,756,382; European Patent 0 394 026; U.S. Patent Publication Nos. 2002/0016332; 2002/0055517; 2002/0110840; 2003/0133913; 2003/0199538; and 2004/0014779; and International Patent Publication Nos. WO 01/74343; WO 02/46749 WO 02/102377; WO 03/020889; WO 03/043572; WO 03/045391; WO 03/103584; and WO 04/058759.
  • Additional examples of small molecule IRMs include certain purine derivatives (such as those described in U.S. Pat. Nos. 6,376,501, and 6,028,076), certain imidazoquinoline amide derivatives (such as those described in U.S. Pat. No. 6,069,149), certain imidazopyridine derivatives (such as those described in U.S. Pat. No. 6,518,265), certain benzimidazole derivatives (such as those described in U.S. Pat. No. 6,387,938), certain derivatives of a 4-aminopyrimidine fused to a five membered nitrogen containing heterocyclic ring (such as adenine derivatives described in U.S. Pat. Nos. 6,376,501; 6,028,076 and 6,329,381; and in International Patent Publication No. WO 02/08905), and certain 3-β-D-ribofuranosylthiazolo[4,5-d]pyrimidine derivatives (such as those described in U.S. Publication No. 2003/0199461).
  • Other IRMs include large biological molecules such as oligonucleotide sequences. Some IRM oligonucleotide sequences contain cytosine-guanine dinucleotides (CpG) and are described, for example, in U.S. Pat. Nos. 6,194,388; 6,207,646; 6,239,116; 6,339,068; and 6,406,705. Some CpG-containing oligonucleotides can include synthetic immunomodulatory structural motifs such as those described, for example, in U.S. Pat. Nos. 6,426,334 and 6,476,000. Other IRM nucleotide sequences lack CpG sequences and are described, for example, in International Patent Publication No. WO 00/75304.
  • Other IRMs include biological molecules such as aminoalkyl glucosaminide phosphates (AGPs) and are described, for example, in U.S. Pat. Nos. 6,113,918; 6,303,347; 6,525,028; and 6,649,172.
  • Any suitable IRM compound may be used to practice the invention. Unless otherwise indicated, reference to a compound can include the compound in any pharmaceutically acceptable form, including any isomer (e.g., diastereomer or enantiomer), salt, solvate, polymorph, and the like. In particular, if a compound is optically active, reference to the compound can include each of the compound's enantiomers as well as racemic mixtures of the enantiomers.
  • In some embodiments, a suitable IRM compound may be, for example, a small molecule IRM compound such as one of those described above. Suitable small molecule IRM compounds include those having a 2-aminopyridine fused to a five membered nitrogen-containing heterocyclic ring such as, for example, imidazoquinoline amines including but not limited to amide substituted imidazoquinoline amines, sulfonamide substituted imidazoquinoline amines, urea substituted imidazoquinoline amines, aryl ether substituted imidazoquinoline amines, heterocyclic ether substituted imidazoquinoline amines, amido ether substituted imidazoquinoline amines, sulfonamido ether substituted imidazoquinoline amines, urea substituted imidazoquinoline ethers, thioether substituted imidazoquinoline amines, and 6-, 7-, 8-, or 9-aryl or heteroaryl substituted imidazoquinoline amines; tetrahydroimidazoquinoline amines including but not limited to amide substituted tetrahydroimidazoquinoline amines, sulfonamide substituted tetrahydroimidazoquinoline amines, urea substituted tetrahydroimidazoquinoline amines, aryl ether substituted tetrahydroimidazoquinoline amines, heterocyclic ether substituted tetrahydroimidazoquinoline amines, amido ether substituted tetrahydroimidazoquinoline amines, sulfonamido ether substituted tetrahydroimidazoquinoline amines, urea substituted tetrahydroimidazoquinoline ethers, and thioether substituted tetrahydroimidazoquinoline amines; imidazopyridine amines including but not limited to amide substituted imidazopyridine amines, sulfonamido substituted imidazopyridine amines, urea substituted imidazopyridine amines, aryl ether substituted imidazopyridine amines, heterocyclic ether substituted imidazopyridine amines, amido ether substituted imidazopyridine amines, sulfonamido ether substituted imidazopyridine amines, urea substituted imidazopyridine ethers, and thioether substituted imidazopyridine amines; 1,2-bridged imidazoquinoline amines; 6,7-fused cycloalkylimidazopyridine amines; imidazonaphthyridine amines; tetrahydroimidazonaphthyridine amines; oxazoloquinoline amines; thiazoloquinoline amines; oxazolopyridine amines; thiazolopyridine amines; oxazolonaphthyridine amines; thiazolonaphthyridine amines; and 1H-imidazo dimers fused to pyridine amines, quinoline amines, tetrahydroquinoline amines, naphthyridine amines, or tetrahydronaphthyridine amines.
  • In certain embodiments, the IRM compound can be a tetrahydroimidazoquinoline amine such as, for example, 4-amino-2-(ethoxymethyl)-α,α-dimethyl-6,7,8,9-tetrahydro-1H-imidazo[4,5-c]quinoline-1-ethanol. In alternative embodiments, the IRM compound may be an imidazoquinoline amine. In certain specific embodiments, the IRM may be 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin4-amine. In other embodiments, the IRM compound may be a sulfonamide substituted imidazoquinoline amine such as, for example, N-[4-(4-amino-2-ethyl-1H-imidazo[4,5-c]quinolin-1-yl)butyl]methanesulfonamide. Various combinations of IRMs can be used if desired.
  • The IRM compound—or each component of a combination such as, for example, an IRM and IL-2 and/or a PKC agonist—may be provided in any formulation suitable for administration to a subject. Suitable types of formulations are described, for example, in U.S. Pat. No. 5,736,553; U.S. Pat. No. 5,238,944; U.S. Pat. No. 5,939,090; U.S. Pat. No. 6,365,166; U.S. Pat. No. 6,245,776; U.S. Pat. No. 6,486,186; European Patent No. EP 0 394 026; and U.S. Patent Publication No. 2003/0199538. The compound—whether an IRM compound, IL-2, or a PKC agonist—may be provided in any suitable form including but not limited to a solution, a suspension, an emulsion, or any form of mixture. The compound may be delivered in formulation with any pharmaceutically acceptable excipient, carrier, or vehicle. For example, the compound may be provided in a formulation suitable for topical administration. Suitable types of formulations for topical delivery of, for example, certain IRM compounds are described, e.g., in International Patent Publication No. WO 03/045391. The formulation may be delivered in any conventional dosage form including but not limited to a cream, an ointment, an aerosol formulation, a non-aerosol spray, a gel, a lotion, a tablet, a lozenge, an elixir, and the like. The formulation may further include one or more additives including but not limited to adjuvants, skin penetration enhancers, colorants, fragrances, moisturizers, thickeners, and the like.
  • The composition of a formulation suitable for practicing the invention will vary according to factors known in the art including but not limited to the physical and chemical nature of the IRM compound, the nature of the carrier, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IRM compound, whether the IRM is being administered in combination with one or more additional agents, and the species to which the formulation is being administered. Accordingly, it is not practical to set forth generally the composition of a formulation effective for all possible applications and all possible embodiments of the invention. Those of ordinary skill in the art, however, can readily determine an appropriate formulation with due consideration of such factors.
  • In some embodiments, the methods of the present invention include administering IRM to a subject in a formulation of, for example, from about 0.0001% to about 10% (unless otherwise indicated, all percentages provided herein are weight/weight with respect to the total formulation) to the subject, although in some embodiments the IRM compound may be administered using a formulation that provides IRM compound in a concentration outside of this range. In certain embodiments, the method includes administering to a subject a formulation that includes from about 0.01% to about 5% IRM compound, for example, a formulation that includes about 5% IRM compound.
  • An amount of an IRM compound effective for practicing the invention will vary according to factors known in the art including but not limited to the physical and chemical nature of the IRM compound, the nature of the carrier, the intended dosing regimen, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IRM compound, whether the IRM is being administered in combination with one or more additional agents, and the species to which the formulation is being administered. Accordingly, it is not practical to set forth generally the amount that constitutes an amount of IRM compound effective for all possible applications and all possible embodiments of the invention. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • In some embodiments, the methods of the present invention include administering sufficient IRM compound to provide a dose of, for example, from about 100 ng/kg to about 50 mg/kg to the subject, although in some embodiments the methods may be performed by administering IRM compound in a dose outside this range. In some of these embodiments, the method includes administering sufficient IRM compound to provide a dose of from about 10 μg/kg to about 5 mg/kg to the subject, for example, a dose of from about 100 μg/kg to about 1 mg/kg.
  • The dosing regimen may depend at least in part on many factors known in the art including but not limited to the physical and chemical nature of the IRM compound, the nature of the carrier, the amount of IRM being administered, the state of the subject's immune system (e.g., suppressed, compromised, stimulated), the method of administering the IRM compound, whether the IRM is being administered in combination with one or more additional agents, and the species to which the formulation is being administered. Accordingly it is not practical to set forth generally the dosing regimen effective for all possible applications and all possible embodiments of the invention. Those of ordinary skill in the art, however, can readily determine an appropriate dosing regimen with due consideration of such factors.
  • In some embodiments of the invention, the IRM compound may be administered, for example, from a single dose to multiple doses per day. In certain embodiments, the IRM compound may be administered from about three times per week to about once per day. In one particular embodiment, the IRM compound is administered once per day.
  • EXAMPLES
  • The following examples have been selected merely to further illustrate features, advantages, and other details of the invention. It is to be expressly understood, however, that while the examples serve this purpose, the particular materials and amounts used as well as other conditions and details are not to be construed in a matter that would unduly limit the scope of this invention.
  • The Compounds used in the examples are shown in Table 1.
    TABLE 1
    Compound Chemical Name Reference
    IRM1 4-amino-2-(ethoxymethyl)-α,α-dimethyl- U.S. Pat No.
    6,7,8,9-tetrahydro-1H-imidazo[4,5- 5,352,784
    c]quinoline-1-ethanol hydrate Example 91
    IRM2 1-(2-methylpropyl)-1H-imidazo[4,5- U.S. Pat. No.
    c]quinolin-4-amine 4,689,338
    Example 99
    IRM3 N-[4-(4-amino-2-ethyl-1H-imidazo[4,5- U.S. Pat. No.
    c]quinolin-1-yl)butyl]methanesulfonamide 6,677,349
    Example 236
    Negative 4-hydroxy-1-isobutyl-1H-imidazo[4,5- Example 71 of
    Control c]quinoline U.S. Pat. No.
    (Neg.) 4,698,348

    Materials and Methods
  • Blood samples: Heparinized blood (30-40 mL) was collected from consenting CLL patients (diagnosed by a persistent elevation of CD19+CD5+IgMlo lymphocytes (Rozman and Montserrat, New Engl. J. Med. 1995;333:1052-1057)). All patients were untreated at the time of analysis. Protocols were approved by the appropriate Institutional Review Board.
    TABLE 2
    Material Commercial Source
    Lipopolysaccharide (LPS) Sigma Chemical Co., St. Louis, MO
    Phorbol dibutyrate (PDB) Sigma Chemical Co., St. Louis, MO
    Clinical grade IL-2 Chiron Corporation, San Francisco, CA
    Interferon-α2b Schering Canada, Pointe-Claire, Quebec
    Dexamethasone Pharmascience, Inc., Montreal, Quebec
    Bryostatin-1 ICN Biomedicals, Inc., Aurora, OH
    Poly (I:C) Amersham Pharmacia Biotech, Inc.,
    Piscataway, NJ
    SB203580 Calbiochem (San Diego, CA
  • Antibodies: Phycoerythrin- or FITC-labeled CD80 (B7-1), CD86 (B7-2), CD54 (ICAM-1), CD83, 4-1BB ligand (4-1BBL), CD5, and CD19 antibodies were purchased from BD Pharmingen (San Francisco, Calif.). Phycoerythrin-labeled ICOS-L and PDL-1 (B7-H1) antibodies were obtained from eBioscience (San Diego, Calif.).
  • Materials Preparation and Methods: Stock solutions of PDB (5 mg/mL) were made in DMSO. Stock solutions of SB203580 (25 mg/mL), an inhibitor of the selective stress-activated protein kinase (SAPK) (p38) (Lee et al., Pharmacol Ther. 1999;82:389-397) were made in DMSO. IRM1 and Negative Control (Neg.) compound were provided by 3M Pharmaceuticals (St. Paul, Minn.). The compounds were dissolved in AIM-V medium (GibcoBRL, Grand Isaland, N.Y.)(with 33% DMSO) at 1.3 mg/mL and stored in the dark at 4° C. A 5% cream of IRM2, marketed as ALDARA, also was provided by 3M Pharmaceuticals.
  • Cell purification: CLL and T cells were isolated from fresh blood by negative selection (RosetteSep, StemCell Technologies, Vancouver, BC) as described by Gitelson et al. (Gitelson et al., Clin. Cancer Res. 2003;99:1656-1665).
  • Activation of CLL cells: Purified CLL cells (1.5×106 cells/mL) were cultured in serum-free AIM-V medium plus 2-mercaptoethanol (2-ME, 5×10−5 M) (Sigma Chemical Co.) in 6- or 24-well plates (Becton-Dickinson Labware, Franklin Lake, N.J.) for 3-4 days at 37° C. in 5% CO2. CLL cells were activated by adding the Negative Control compound (1 μg/mL), IRM1 (1 μg/mL), IL-2 (5000 U/mL), PDB (100 ng/mL), or bryostatin (20 nmol), as appropriate. The Negative Control compound did not have measurable effects on CLL cells and, consequently, AIM-V medium, alone, was used as a control for the majority of the experiments.
  • Mixed Lymphocyte Responses (MLRs): T cells were isolated from CLL patients and adjusted to 5×105 cells/mL in AIM-V medium. Activated CLL cells were washed at least 4 times to remove residual immunomodulators, irradiated (2500 cGy) and suspended at 5×105 cells/mL (or lower concentrations) in AIM-V. Responders and stimulators were then mixed in a 1:1 (vol:vol) ratio and cultured in 96-well round bottom plates (Becton Dickinson Labware, Franklin Lake, N.J.) without additional cytokines or serum. Proliferation was measured 4-6 days later using a colorimetric assay (Gitelson et al., Clin Cancer Res. 2003;99: 1656-1665; and Ahmed et al., J. Immunol. Methods. 1994; 170:211-224).
  • Flow cytometry: Cell staining was performed as described by Gitelson et al. (Gitelson et al., Clin. Cancer Res. 2003;99:1656-1665).
  • Cytokine measurement: Cytokine levels in culture supernatants (from activated CLL cells after 48 hours) were determined by a multi-analysis fluorescent bead assay system available from Luminex Corp., Austin, Tex., under the tradename LUMINEX-100 SYTEM. A 5-plex human cytokine kit for IFN-γ, IL-2, IL-4, IL-10 and TNF-α measurement was used, according to the manufacturer's instructions (R&D Systems, Minneapolis, Minn.). Individual cytokine concentrations were determined from standard curves using software available from BioRad, Mississauga, Ontario, under the tradename BIO-PLEX 2.0. The assay was linear between 30 and 1000 μg/mL for each cytokine.
  • Statistical analysis: The Student t-test was used to determine p-values for differences between sample means. Best-fit lines were determined by least-squares regression.
  • Example 1 Effect of IRM1 on Costimulatory Molecule Expression by CLL Cells
  • CLL cells from the indicated number of patients were cultured in IRM1 (1 μg/mL) for 3-4 days, and then assayed for expression of the costimulatory molecules indicated on the x-axis (at an intensity greater than the first decade of log fluorescence) of FIG. 1A. The percentage of cells that expressed each costimulatory molecule and the mean fluorescence intensity (MFI) of expression were measured by flow cytometry. The “fold-increase” was then calculated from the ratio of these measurements to the percentage and MFI of control cells cultured without activating agents. The average and standard error of these relative increases in costimulatory molecule expression are shown in the FIG. 1A.
  • IRM1 has especially strong effects on CD80, CD86, and CD54 expression, with IRM1 increasing the expression of CD54, CD80, and CD86 on CLL cells from all patients studied (n=31). The effect on CD80 was greater than on CD86 (compare FIGS. 1, 2). IRM1 also increased the expression of CD83, 4-1BBL, and PDL-1, but had little effect on ICOS-L expression (FIG. 1A).
  • CLL cells from the indicated number of patients were cultured in IRM1, LPS (100 μg/mL), poly (I:C) (100 μg/mL) and IFN-γ 2B (500 U/mL) for 48 hours. Relative increases in the expression of CD80 and CD86 were calculated as described for FIG. 1A, and are shown in FIG. 1B.
  • CLL cells were not affected in the same way by other TLR agonists. TLR2 and TLR4 are activated by bacterial LPSs while TLR3 is activated by viral double-stranded RNA and poly (I:C) (Gordon, Cell. 2002;111:927-930). However LPS or poly (I:C) rarely affected costimulatory molecule expression by CLL cells (FIG. 1B). Although IRM1 is one of a class of IRMs known to stimulate the release of IFN-α from DCs or monocytes (Gibson et al., Cell Immunol. 2002;218:74-86), the effects of IRM1 were unlikely to be mediated indirectly by this cytokine since costimulatory molecule expression by CLL cells did not change significantly after direct stimulation with IFN-γ2B (FIG. 1B).
  • Example 2 Effects of IL-2 and IRM1 on Costimulatory Molecule Expression by CLL Cells
  • CLL cells were isolated and cultured alone or with IL-2 (5000 U/mL), IRM1 (1 μg/mL), or both IL-2 and IRM1 for 3-4 days. The expression of CD80, CD86, CD54, and CD83 was then determined by flow cytometry. FIG. 2A shows a characteristic example. The numbers in the dot plots in the upper and lower rows are the percentages of CD80+ and CD86+ CLL cells, respectively. FIG. 2B is a graphical representation of the percentage of CLL cells expressing the different costimulatory molecules (determined by the percentage of cells with staining intensity above the first decade of log fluorescence) from the number of patients indicated in the graph legend. The average and standard error for each of these measurements are shown in the graphs. The numbers over the double-headed arrows represent the p-values for the differences between sample means. FIG. 2C is a graphical representation of the mean fluorescence index (MFI) of expression of the different costimulatory molecules determined for CLL cells from the number of patients indicated in the graph legend. The average and standard error for each of these measurements are shown in the graphs. Only the MFI of CD54 expression (divided by 10) is shown since essentially all CLL cells express this molecule. The numbers over the double-headed arrows represent the p-values for the differences between sample means.
  • IL-2 and IRM1 both increased the percentage of CLL cells that expressed CD80 and CD86, as well as the mean fluorescence intensity (MFI) of expression of these molecules (FIG. 2). As a single agent, IRM1 appeared to be more potent than IL-2 in this regard. The effects of IL-2 and IRM1 on costimulatory molecule expression were additive (FIG. 2A, right dot-plots and FIG. 2B,C), suggesting they were mediated by different mechanisms. The MFI for the costimulatory molecules CD80, CD86, CD54, and CD83 shown in FIG. 2C indicate that IRM1 increased the expression of all four of these costimulatory molecules on CLL cells. The magnitude of CD80 expression was especially increased by IRM 1 in combination with IL-2.
  • Expression of 4-1 BBL and PDL-1 was increased somewhat by IL-2 and IRM1, but not as much as CD80, CD86, and CD54 (FIG. 1A). ICOS-L was found on many CLL cells but its expression appeared to be relatively independent of IL-2 and IRM 1-mediated signaling.
  • Example 3 Effect of PKC Activation on Costimulatory Phenotype and T Cell Stimulatory Ability of IL-2- and IRM1-Activated CLL Cells
  • CLL cells were purified from individual patients and cultured alone or with IL-2, IRM1, IL-2 and IRM1, PDB, PDB and IL-2, PDB and IRM1, or PDB, IL-2, and IRM1. After 3-4 days, these cells were harvested, washed extensively, irradiated (2500 cGy) and used to stimulate allogenic or autologous T cells from CLL patients (obtained at the same time as the CLL cells and rested in culture until added to the Mixed Lymphocytic Response (MLR) assay) after 5-6 days of culture, alamar blue was added and proliferation was measured in an optical density colorimetric microplate reader at wavelengths of 540 (reduced state) and 595 (oxidized state). The difference between these readings was used as a measure of the number of viable cells in the culture. Results are shown in FIG. 3A. The results from the T cell source that exhibited the greatest stimulation (after subtraction of the proliferation induced by non-activated CLL cell stimulators) from each individual experiment were used to generate the average proliferation and standard error from the number of patients indicated on the x-axis.
  • FIG. 3B is a graphical representation of the correlation of CD83 expression with T cell stimulatory ability. CLL cells were treated with IL-2 and IRM1 for 4-5 days. The percentage of cells expressing CD83 and the MFI of CD83 expression were then determined by flow cytometry. The activated CLL cells were then irradiated and used to stimulate autologous or allogeneic T cells in MLRs. The initial percentages of CD83+ CLL cells (left panel) and the MFI of CD83 expression (right panel) from 19 different patients were then correlated with the measured proliferation in the MLRs. The best straight line has intercept 10.692 and slope 0.0598; the associated P-value is 0.0153.
  • FIG. 4A shows CLL cells from a representative patient that were cultured alone (left panels) or with IRM1, IL-2, and PDB (right panels) for 3 days. CD80, CD83, CD54, and CD86 expression were then determined by flow cytometry. The percentages in the dot-plots refer to CD80 (sum of the right and left upper quadrants) (top panels) and to CD86 (sum of the right upper and lower quadrants) (bottom panels). FIG. 4B is a summary of the results of flow cytometric evaluation of the percentage of CLL cells expressing CD80, CD83, CD54, and CD86 (and the MFI of expression) after culture alone, with PDB, PDB and IL-2, PDB and IRM1, or PDB, IL-2 and IRM1 from the indicated number of patients in the graph legend. The average and standard error are shown. Only the MFI of CD54 expression is shown since essentially all CLL cells express this molecule. FIG. 4C is a summary of similar flow cytometric evaluation of ICOS-L, 4-1BBL, and PDL-1 expression. The numbers over the double-headed arrows are the p-values for the differences between sample means.
  • Treatment with phorbol dibutyrate (PDB), alone, caused ˜90% of CLL cells to express CD83 (FIG. 4B, clear bars). PDB also increased the number of CD80+ and CD86+ CLL cells (the latter more than the former), as well as the expression of 4-1BBL and PDL-1 (FIG. 4C, clear bars). CD54 and ICOS-L expression were not affected greatly by PDB (FIG. 4B and FIG. 4C).
  • Addition of IL-2 during activation of CLL cells with PDB increased mainly the number of CD80+ cells and the MFI of CD80 and CD54 expression (FIG. 4B; horizontal bars). A slightly greater percentage of CD80+ cells was obtained when CLL cells were activated with both PDB and IRM1 (FIG. 4B; vertical bars). The addition of IL-2 to IRM1 and PDB strongly increased the expression of CD80 (especially compared to CD86 (FIG. 4B)), as well as CD54, and caused essentially all CLL cells to acquire a CD83hiCD80hiCD86hiCD54hi cell surface phenotype (FIG. 4A and FIG. 4B; diagonal bars).
  • The results shown in FIG. 4 indicated that the combination of PDB and IRM1 caused nearly 100% of CLL cells to acquire CD80, CD86, and CD83 expression. The addition of IL-2 affected mainly the magnitude of CD80 and CD54 expression. PDB, with or without IL-2, and/or IRM1 increased the expression of 4-1BBL and PDL-1 but not to the same extent as CD80, CD86, CD54, and CD83.
  • This strong expression of costimulatory molecules by CLL cells activated with PDB, IL-2, and IRM1 was reflected in the ability of these cells to stimulate T cell proliferation (FIG. 3A). CLL cells stimulated with PDB (without IL-2) were weak stimulators of T cell proliferation (FIG. 3A).
  • Example 4 Elimination of CLL Cells by Autologous T Cells in the Presence of IRM1
  • CLL cells and T cells were isolated from a CLL patient, suspended at concentrations of 106 cells/mL and mixed in a 1:1 ratio. The cell mixtures were cultured alone, or in the presence of IL-2, IL-2 and IRM1, bryostatin, bryostatin and IL-2, bryostatin and IRM1, or bryostatin, IL-2, and IRM1. In FIG. 5A, after 5 days, the percentages of CD5+ CD19+ tumor cells (indicated by the numbers in the right upper quadrants of the dot-plots) and CD5+ CD19 T cells were determined by flow cytometry. In FIG. 5B, these percentages and the total numbers of viable cells (determined by manual counting in a hemocytometer) were used to calculate the remaining absolute numbers of CLL cells in the cultures.
  • IRM1, whether alone or in combination with IL-2 and/or bryostatin, induced autologous T cells to kill CLL cells in vitro. The combination of IRM1, IL-2, and bryostatin enabled autologous T cells to achieve 100% clearance of CLL cells in 5 days.
  • Example 5 Clinical Effects of Administering an IRM Compound to Lymphomatous Skin Deposits Associated with Chronic Lymphocytic Leukemia
  • A 71-year old Caucasian male was diagnosed with Rai Stage 0 CLL on the basis of a persistent elevated count of circulating monoclonal CD19+CD5+IgMlo lymphocytes, determined by flow cytometry. CD38 was expressed by 45% of circulating CLL cells. The white blood cell counts at the beginning and end of treatment with IRM2 were 36×106 cells/mL and 45×106 cells/mL, respectively. Other systemic chemotherapy, steroids, or radiation had not been administered previously.
  • Additionally, the patient reported having recurrent nodular, erythematous lesions on his hands and arms for approximately eight years. The lesions were usually removed by treatment with liquid nitrogen. At the time he was diagnosed with CLL, he had several such lesions over his upper back (FIG. 8A) and arms. One lesion was biopsied and found to contain a diffuse atypical lymphoid dermal infiltrate consisting of many small, round lymphocytes, without epidermotropism. On paraffin immunoperoxidase stains, the lymphoid filtrate had a predominant CD20+ phenotype. Molecular analysis on paraffin embedded tissue demonstrated a monoclonal B cell population, consistent with B cell lymphoma.
  • A 5% cream of IRM2 was applied to the affected area three times per week. After eight weeks, the size of the treated lesion had not changed significantly, although an area of hypopigmentation had formed that was reminiscent of a halo nevus around a regressing melanoma deposit.
  • Administration of 5% IRM2 cream was increased to once per day. The lesion disappeared after six weeks of treatment (FIG. 6B) and had not recurred by three months after treatment ceased. Neither untreated lymphomatous lesions nor circulating white blood cell count changed significantly over the course of the treatment.
  • CLL cells were isolated from fresh blood by negative selection (RosetteSep, StemCell Technologies, Inc., Vancouver, BC) as described in Gitelson et al., Clin. Can. Res., 9:1656-1665 (2003). Purified CLL cells (1.5×106 cells/mL) were cultured in serum-free AIM-V medium (GibcoBRL) for three days. IRM1 and Negative Control compound were used at final concentrations of 1 μg/mL.
  • Cells were incubated with pre-optimized volumes of either CD80-PE and CD83-FITC or CD54-PE and CD86-FITC antibodies for 20 minutes, washed, then subjected to flow cytometry analysis. Negative controls were isotype-matched irrelevant antibodies. Staining of nucleated cells was determined by gating on forward- and side-scatter properties. Ten thousand viable counts were analyzed with a FACScan flow cytometer using CELLQUEST software (BD Immunocytometry Systems, San Jose, Calif.). The flow cytometer was standardized with SpheroParticles (Spherotech, Inc., Chicago, Ill.). Percentages of CD80+, CD86+, and CD83+ cells were calculated by comparison with isotype control-labeled cells. Results are shown in FIG. 7.
  • Example 6 Dose Response of IRM3 Effects on Costimulatory Marker Expression by CLL Cells
  • CLL cells from eight different patients were purified and cultured with either 0.001 μg/mL, 0.01 μg/mL, 0.1 μg/mL, or 1.0 μg/mL of IRM3 for three days. CD80 and CD83 expression was determined by flow cytometry as described above. Increases in CD80 and CD83 expression were computed by subtracting CD80 and CD83 expression, respectively, from control CLL cultures to which no IRM3 was added. Results are presented in FIG. 8.
  • EXAMPLE 7 IRM3-Mediated Changes in CLL Cell Surface Molecules
  • CLL cells were collected from patients and cultured for 2-3 days with IRM3 (1 μg/mL) and without (control). The percentages of cells that expressed CD80, CD83, CD86, and CD38 were determined by flow cytometry. The MFI of expression of CD54 and CD25 was determined because virtually all CLL cells express these molecules. Changes in MFI of expression or the percentage of cells expressing a particular cell surface molecule were determined by subtracting values obtained from control cultures from the values obtained from cells cultured with IRM3. Results are presented in FIG. 9.
  • Example 8 IRM3-Mediated Increase in Expression of CD20 by CLL Cells
  • CLL cells were purified and cultured either without (control) or with IRM3 (1 μg/mL) and IL-2 (5000 U/mL). After 48 hours, the MFI of CD20 expression was determined by flow cytometry. Results are presented in FIG. 10.
  • The complete disclosures of the patents, patent documents and publications cited herein are incorporated by reference in their entirety as if each were individually incorporated. In case of conflict, the present specification, including definitions, shall control. Various modifications and alterations to this invention will become apparent to those skilled in the art without departing from the scope and spirit of this invention. Illustrative embodiments and examples are provided as examples only and are not intended to limit the scope of the present invention. The scope of the invention is limited only by the claims set forth as follows.

Claims (67)

1. A method of increasing the expression of at least one cell surface molecule of CD5+ B cell lymphoma cells, the method comprising contacting the CD5+ B cell lymphoma cells with at least one IRM compound effective to increase the expression of at least one cell surface molecule of the CD5+ B cell lymphoma cells.
2. The method of claim 1 wherein the CD5+ B cell lymphoma cells are selected from the group consisting of chronic lymphocytic leukemia (CLL) cells, small lymphocytic lymphoma cells (SLL), mantle cell lymphoma cells, splenic lymphoma with villous lymphocytes, and combinations thereof.
3. The method of claim 1 wherein the IRM is a TLR7 agonist.
4. The method of claim 1 wherein the IRM is a TLR8 agonist.
5. The method of claim 1 wherein the IRM is a tetrahydroimidazoquinoline amine.
6. The method of claim 1 wherein the IRM is a sulfonamide substituted imidazoquinoline amine.
7. The method of claim 1 wherein the cell surface molecule is a costimulatory molecule.
8. The method of claim 7 wherein at least one costimulatory molecule is CD25, CD38, CD40, CD54, CD80, CD83, or CD86.
9. The method of claim 1 wherein the cell surface molecule is CD20, CD22, or CD23.
10. The method of claim 1 wherein contacting the CD5+ B cell lymphoma cells with an IRM occurs in vitro.
11. The method of claim 1 wherein contacting the CD5+ B cell lymphoma cells with an IRM occurs in an organ, tissue, or blood.
12. The method of claim 1 wherein contacting the CD5+ B cell lymphoma cells with an IRM occurs in vivo in a subject.
13. A method of stimulating CD5+ B cell lymphoma cells to produce a cytokine, the method comprising contacting the CD5+ B cell lymphoma cells with an IRM effective to produce at least one cytokine above a level produced by the CD5+ B cell lymphoma cells not contacted by the IRM, wherein the at least one cytokine is IL-1β, IL-6, IL-8, IL-10, IL-12, TNF-α, GM-CSF, or a combination thereof.
14. The method of claim 13 wherein contacting the CD5+ B cell lymphoma cells occurs in vitro.
15. The method of claim 13 wherein contacting the CD5+ B cell lymphoma cells occurs in an organ, tissue, or blood.
16. The method of claim 13 wherein contacting the CD5+ B cell lymphoma cells occurs in vivo in a subject.
17. A method of increasing T cell proliferation, the method comprising:
contacting CD5+ B cell lymphoma cells with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of CD5+ B cell lymphoma cells; and
contacting the CD5+ B cell lymphoma cells with T cells, thereby activating the T cells;
wherein the activated T cells demonstrate increased proliferation compared to T cells contacted with CD5+ B cell lymphoma cells that have not been contacted with an IRM.
18. The method of claim 17, further comprising contacting the CD5+ B cell lymphoma cells with at least one additional immunomodulating agent.
19. The method of claim 18 wherein the additional immunomodulating agent comprises IL-2.
20. The method of claim 18 wherein the additional immunomodulating agent comprises a protein kinase C agonist.
21. The method of claim 18 wherein the T cells and the CD5+ B cell lymphoma cells are contacted in vivo.
22. A method of increasing the killing of CD5+ B cell lymphoma cells by CD5+ B cell lymphoma cell-specific cytotoxic T cells, the method comprising:
contacting CD5+ B cell lymphoma cells with an IRM effective to increase the expression of at least one costimulatory molecule on the surface of the CD5+ lymphoma cells; and
contacting the CD5+ lymphoma cells with CD8+ T cells, thereby activating the CD8+ T cells;
wherein the activated CD8+ T cells are CD5+ B cell lymphoma cell-specific cytotoxic T cells that demonstrate increased killing of CD5+ B cell lymphoma cells compared to CD8+ T cells contacted with CD5+ B cell lymphoma cells that have not been contacted with an IRM.
23. The method of claim 22, further comprising contacting the CD5+ B cell lymphoma cells with at least one additional immunomodulating agent.
24. The method of claim 23 wherein the additional immunomodulating agent comprises IL-2.
25. The method of claim 23 wherein the additional immunomodulating agent comprises a protein kinase C agonist.
26. The method of claim 22 wherein the CD5+ B cell lymphoma cells are contacted with the IRM in vivo.
27. A method of increasing the killing of CD5+ B cell lymphoma cells by autologous T cells in a subject suffering from a CD5+ B cell lymphoma, the method comprising administering to the subject an IRM that is a TLR7 and/or TLR8 agonist.
28. The method of claim 27 further comprising administering IL-2.
29. The method of claim 27 further comprising administering a protein kinase C agonist.
30. The method of claim 27 further comprising administering IL-2 and a protein kinase C agonist.
31. A method of treating a subject suffering from a CD5+ B cell lymphoma, the method comprising administering to the subject an IRM effective to increase the expression of at least one cell surface molecule of the CD5+ B cell lymphoma cells in an amount effective to increase the expression of at least one cell surface molecule of the CD5+ B cell lymphoma cells.
32. The method of claim 31 wherein the IRM is a TLR7 and/or TLR8 agonist.
33. The method of claim 31 further comprising administering IL-2.
34. The method of claim 31 further comprising administering a protein kinase C agonist.
35. The method of claim 31 further comprising administering IL-2 and a protein kinase C agonist.
36. The method of claim 31 wherein at least one cell surface molecule whose expression is increased is a target of a therapeutic agent, and the method further comprises administering to the subject an effective amount of the therapeutic agent.
37. The method of claim 26 wherein the cell surface molecule is CD20, CD22, or CD23.
38. The method of claim 31 wherein the cell surface molecule is a costimualtory molecule.
39. A method of treating a CD5+ B cell lymphoma, the method comprising administering to a subject in need of such treatment an IRM effective to ameliorate at least one symptom or clinical sign characteristic of a the CD5+ B cell lymphoma.
40. The method of claim 39 wherein the IRM is administered in an amount effective to demonstrate at least a 50% decrease in peripheral blood lymphocytes, lymphadenopathy, or splenomegaly for at least two months.
41. The method of claim 39 wherein the IRM is administered in an amount effective to prevent the development of progressive disease, wherein progressive disease is at least a 50% increase in circulating lymphocytes or a progression to a more aggressive histology.
42. The method of claim 39 wherein the IRM is administered in an amount effective to ameliorate erythematous lesions.
43. A vaccine comprising isolated CD5+ B cell lymphoma cells or an immunologically active portion thereof, wherein isolated CD5+ B cell lymphoma cells have been contacted with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of the CD5+ B cell lymphoma cells.
44. The vaccine of claim 43 wherein the IRM is a TLR7 and/or TLR8 agonist.
45. The vaccine of claim 43 wherein the isolated CD5+ B cell lymphoma cells have further been contacted with IL-2.
46. The vaccine of claim 43 wherein the isolated CD5+ B cell lymphoma cells have further been contacted with a protein kinase C agonist.
47. The vaccine of claim 43 wherein the isolated CD5+ B cell lymphoma cells have further been contacted with IL-2 and a protein kinase C agonist.
48. A method of preparing a vaccine comprising contacting isolated CD5+ B cell lymphoma cells with an IRM effective to increase the expression of at least one molecule on the surface of the CD5+ B cell lymphoma cells.
49. The method of claim 48 further comprising contacting said isolated cells with IL-2.
50. The method of claim 48 further comprising contacting said isolated cells with a protein kinase C agonist.
51. The method of claim 48 further comprising contacting said isolated cells with IL-2 and a protein kinase C agonist.
52. A method of treating a subject suffering from a CD5+ lymphoma, the method comprising administering to the subject an immunologically active portion of isolated CD5+ B cell lymphoma cells, wherein the isolated CD5+ B cell lymphoma cells have been contacted with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of the CD5+ B cell lymphoma cells.
53. The method of claim 52 wherein the isolated CD5+ B cell lymphoma cells have also been contacted with IL-2.
54. The method of claim 52 wherein the isolated the CD5+ B cell lymphoma cells have also been contacted with a protein kinase C agonist.
55. The method of claim 52 wherein the isolated CD5+ B cell lymphoma cells have also been contacted with IL-2 and a protein kinase C agonist.
56. The method of claim 52 wherein the isolated CD5+ B cell lymphoma cells are derived from the subject suffering from a CD5+ B cell lymphoma.
57. The method of claim 52 wherein the immunologically active portion of isolated CD5+ B cell lymphoma cells comprises whole cells.
58. The method of claim 52 wherein the immunologically active portion of isolated CD5+ B cell lymphoma cells comprises a cell membrane preparation or a protein preparation from the isolated CD5+ B cell lymphoma cells.
59. The method of claim 52 wherein at least one cell surface molecule whose expression is increased is a target of a therapeutic agent, and the method further comprises administering to the subject an effective amount of the therapeutic agent.
60. A method of initiating a CD5+ B cell lymphoma-reactive T cell response in a subject diagnosed as having a CD5+ B cell lymphoma, the method comprising administering to the subject a composition comprising an immunologically active portion of isolated CD5+ B cell lymphoma cells that have been contacted with an IRM effective to increase the expression of at least one costimulatory molecule on the cell surface of the CD5+ B cell lymphoma cells.
61. The method of claim 60 wherein the isolated CD5+ B cell lymphoma cells have also been contacted with IL-2.
62. The method of claim 60 wherein the isolated CD5+ B cell lymphoma cells have also been contacted with a protein kinase C agonist.
63. The method of claim 60 wherein the isolated CD5+ B cell lymphoma cells have also been contacted with IL-2 and a protein kinase C agonist.
64. The method of claim 60 wherein the isolated CD5+ B cell lymphoma cells are derived from the subject suffering from the CD5+ lymphoma.
65. The method of claim 60 wherein the isolated CD5+ B cell lymphoma cells are derived from a subject suffering from CLL or SLL.
66. The method of claim 60 wherein the immunologically active portion of isolated CD5+ B cell lymphoma cells comprises whole cells.
67. The method of claim 60 wherein the immunologically active portion of isolated CD5+ B cell lymphoma cells comprises a cell membrane preparation or a protein preparation from the isolated CD5+ B cell lymphoma cells.
US10/933,594 2003-09-05 2004-09-03 Treatment for CD5+ B cell lymphoma Abandoned US20050054665A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/933,594 US20050054665A1 (en) 2003-09-05 2004-09-03 Treatment for CD5+ B cell lymphoma
US12/173,719 US7923429B2 (en) 2003-09-05 2008-07-15 Treatment for CD5+ B cell lymphoma

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US50047803P 2003-09-05 2003-09-05
US56144004P 2004-04-12 2004-04-12
US10/933,594 US20050054665A1 (en) 2003-09-05 2004-09-03 Treatment for CD5+ B cell lymphoma

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/173,719 Continuation US7923429B2 (en) 2003-09-05 2008-07-15 Treatment for CD5+ B cell lymphoma

Publications (1)

Publication Number Publication Date
US20050054665A1 true US20050054665A1 (en) 2005-03-10

Family

ID=34278707

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/933,594 Abandoned US20050054665A1 (en) 2003-09-05 2004-09-03 Treatment for CD5+ B cell lymphoma
US12/173,719 Expired - Fee Related US7923429B2 (en) 2003-09-05 2008-07-15 Treatment for CD5+ B cell lymphoma

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/173,719 Expired - Fee Related US7923429B2 (en) 2003-09-05 2008-07-15 Treatment for CD5+ B cell lymphoma

Country Status (6)

Country Link
US (2) US20050054665A1 (en)
EP (1) EP1660026A4 (en)
JP (1) JP2007504269A (en)
AU (1) AU2004270201A1 (en)
CA (1) CA2537763A1 (en)
WO (1) WO2005023190A2 (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040214851A1 (en) * 2003-04-28 2004-10-28 3M Innovative Properties Company Compositions and methods for induction of opioid receptors
US20070060754A1 (en) * 2003-10-03 2007-03-15 Lindstrom Kyle J Alkoxy substituted imidazoquinolines
US20070066639A1 (en) * 2003-08-12 2007-03-22 Kshirsagar Tushar A Oxime substituted imidazoquinolines
US20070072893A1 (en) * 2003-11-25 2007-03-29 Krepski Larry R Substituted imidazo ring systems and methods
US20070208052A1 (en) * 2004-06-18 2007-09-06 Prince Ryan B Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US20070213356A1 (en) * 2004-06-15 2007-09-13 Merrill Bryon A Nitrogen-Containing Heterocyclyl Substituted Imidazoquinolines and Imidazonaphthyridines
US20070219196A1 (en) * 2004-03-24 2007-09-20 Krepski Larry R Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US20070219228A1 (en) * 2004-06-18 2007-09-20 Shri Niwas Aryl substituted imidazonaphthyridines
US20070287725A1 (en) * 2004-06-18 2007-12-13 3M Innovative Properties Company Isoxazole, Dihydroisoxazole, And Oxadiazole Substituted Imidazo Ring Compounds And Method
US20080070907A1 (en) * 2006-07-12 2008-03-20 Coley Pharmaceutical Group, Inc. Substituted chiral fused [1,2] imidazo [4,5-C] ring compounds and methods
US20080193474A1 (en) * 2005-04-25 2008-08-14 Griesgraber George W Immunostimulatory Compositions
US20080269192A1 (en) * 2004-12-30 2008-10-30 Coley Pharmaceutical Group, Inc. Chiral Fused [1,2]Imidazo[4,5-C] Ring Compounds
US20080306252A1 (en) * 2000-12-08 2008-12-11 Coley Pharmaceutical Group, Inc. Sulfonamido ether substituted imidazoquinolines
US20090018122A1 (en) * 2003-08-27 2009-01-15 Lindstrom Kyle J Aryloxy and Arylalkyleneoxy Substituted Imidazoquinolines
US20090017076A1 (en) * 2003-09-05 2009-01-15 Coley Pharmaceutical Group, Inc. Treatment for cd5+ b cell lymphoma
US20090030030A1 (en) * 2003-12-29 2009-01-29 Bonk Jason D Arylalkenyl and arylalkynyl substituted imidazoquinolines
US20090062272A1 (en) * 2003-12-30 2009-03-05 Bonk Jason D Imidazoquinolinyl sulfonamides
US20090075980A1 (en) * 2003-10-03 2009-03-19 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and Analogs Thereof
US20090105295A1 (en) * 2003-11-14 2009-04-23 Coley Pharmaceutical Group, Inc. Hydroxylamine substituted imidazoquinolines
CN101460178A (en) * 2006-04-07 2009-06-17 艾德拉药物股份有限公司 Stabilized immune modulatory RNA (SIMRA) compounds for TLR7 and TLR8
US20090163532A1 (en) * 2005-02-04 2009-06-25 Coley Pharmaceutical Group, Inc. Aqueous Gel Formulations Containing Immune Response Modifiers
US20090270443A1 (en) * 2004-09-02 2009-10-29 Doris Stoermer 1-amino imidazo-containing compounds and methods
US20090318435A1 (en) * 2003-10-03 2009-12-24 Hays David S Pyrazolopyridines and analogs thereof
US7897767B2 (en) 2003-11-14 2011-03-01 3M Innovative Properties Company Oxime substituted imidazoquinolines
US20110070575A1 (en) * 2004-12-08 2011-03-24 Coley Pharmaceutical Group, Inc. Immunomodulatory Compositions, Combinations and Methods
US7943636B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
US20110117204A1 (en) * 2005-01-28 2011-05-19 Galenbio, Inc. Immunologically active compositions
US7968563B2 (en) 2005-02-11 2011-06-28 3M Innovative Properties Company Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040265351A1 (en) 2003-04-10 2004-12-30 Miller Richard L. Methods and compositions for enhancing immune response
NZ546274A (en) 2003-10-03 2009-12-24 3M Innovative Properties Co Pyrazolopyridines and analags thereof
US8541438B2 (en) 2004-06-18 2013-09-24 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
AU2006338521A1 (en) 2005-02-09 2007-10-11 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted thiazolo(4,5-c) ring compounds and methods
AU2006212765B2 (en) 2005-02-09 2012-02-02 3M Innovative Properties Company Alkyloxy substituted thiazoloquinolines and thiazolonaphthyridines
CA2597446A1 (en) * 2005-02-11 2006-08-31 Coley Pharmaceutical Group, Inc. Substituted imidazoquinolines and imidazonaphthyridines
AU2006216686A1 (en) * 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Method of preferentially inducing the biosynthesis of interferon
JP2008531567A (en) * 2005-02-23 2008-08-14 コーリー ファーマシューティカル グループ,インコーポレイテッド Hydroxyalkyl-substituted imidazoquinoline compounds and methods
US8178677B2 (en) * 2005-02-23 2012-05-15 3M Innovative Properties Company Hydroxyalkyl substituted imidazoquinolines
ZA200803029B (en) 2005-09-09 2009-02-25 Coley Pharm Group Inc Amide and carbamate derivatives of alkyl substituted /V-[4-(4-amino-1H-imidazo[4,5-c] quinolin-1-yl)butyl] methane-sulfonamides and methods
EA200800782A1 (en) * 2005-09-09 2008-08-29 Коли Фармасьютикал Груп, Инк. AMIDA AND CARBAMATE DERIVATIVES N- {2- [4-AMINO-2- (ETOXIMETHYL) -1H-IMIDAZOLO [4,5-c] QUINOLIN-1-IL] -1,1-DIMETHYLETHYL} METHANE SULFONAMIDE AND METHODS
JP5247458B2 (en) 2005-11-04 2013-07-24 スリーエム・イノベイティブ・プロパティーズ・カンパニー Hydroxy and alkoxy substituted 1H-imidazoquinolines and methods
EP1968582A4 (en) * 2005-12-28 2011-02-16 3M Innovative Properties Co Treatment for cutaneous t cell lymphoma
WO2007100634A2 (en) 2006-02-22 2007-09-07 3M Innovative Properties Company Immune response modifier conjugates
WO2007106854A2 (en) * 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Hydroxy and alkoxy substituted 1h-imidazonaphthyridines and methods
US8178539B2 (en) * 2006-09-06 2012-05-15 3M Innovative Properties Company Substituted 3,4,6,7-tetrahydro-5H-1,2a,4a,8-tetraazacyclopenta[cd]phenalenes and methods
US20100160368A1 (en) 2008-08-18 2010-06-24 Gregory Jefferson J Methods of Treating Dermatological Disorders and Inducing Interferon Biosynthesis With Shorter Durations of Imiquimod Therapy
CA3033133C (en) 2009-03-25 2021-11-09 The Board Of Regents Of The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US20110207766A1 (en) 2009-07-13 2011-08-25 Graceway Pharmaceuticals, Llc. Lower dosage strength imiquimod formulations and short dosing regimens for treating genital and perianal warts
HUE033901T2 (en) 2010-08-17 2018-01-29 3M Innovative Properties Co Lipidated immune response modifier compound compositions, formulations, and methods
JP6460789B2 (en) 2011-06-03 2019-01-30 スリーエム イノベイティブ プロパティズ カンパニー Heterobifunctional linker having polyethylene glycol segment and immune response modulating complex prepared from the linker
BR112013031039B1 (en) 2011-06-03 2020-04-28 3M Innovative Properties Co hydrazine compounds 1h-imidazoquinoline-4-amines, conjugates made from these compounds, composition and pharmaceutical composition comprising said compounds and conjugates, uses thereof and method of manufacturing the conjugate
US10286065B2 (en) 2014-09-19 2019-05-14 Board Of Regents, The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds
WO2017214285A1 (en) 2016-06-07 2017-12-14 Eynav Klechevsky Detection of cd5 and methods and compositions for modulating cd5
EP3728255B1 (en) 2017-12-20 2022-01-26 3M Innovative Properties Company Amide substituted imidazo[4,5-c]quinoline compounds with a branched chain linking group for use as an immune response modifier

Citations (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10007A (en) * 1853-09-13 Gear op variable cut-ofp valves for steau-ehgietes
US14779A (en) * 1856-04-29 Xwrench
US22302A (en) * 1858-12-14 Machine for turning irregular forms
US23870A (en) * 1859-05-03 Island
US55517A (en) * 1866-06-12 Improvement in hand corn - planters
US91491A (en) * 1869-06-15 Improved ash-sifter
US110840A (en) * 1871-01-10 Improvement in shoes
US130299A (en) * 1872-08-06 Improvement in fruit-baskets
US132079A (en) * 1872-10-08 Improvement in nut-locks
US133913A (en) * 1872-12-10 Improvement in car-brakes
US139364A (en) * 1873-05-27 Improvement in carriage-protectors
US141950A (en) * 1873-08-19 Improvement in stoppers for bottles
US144283A (en) * 1873-11-04 Improvement in match-boxes
US147543A (en) * 1874-02-17 Improvement in piston-packings
US161797A (en) * 1875-04-06 Improvement in combined water-coolers and refrigerators
US162309A (en) * 1875-04-20 Improvement in gas-retorts
US162332A (en) * 1875-04-20 Improvement in machines for coating fabrics
US171086A (en) * 1875-12-14 Improvement in throttle-valves
US175336A (en) * 1876-03-28 Improvement in grand-piano-forte frames
US176367A (en) * 1876-04-18 Improvement in safety-locks for fire-arms
US180919A (en) * 1876-08-08 Improvement in clothes-sticks
US181130A (en) * 1876-08-15 Improvement in butter-packages
US181211A (en) * 1876-08-15 Improvement in cotton chopper and scraper
US191833A (en) * 1877-06-12 Improvement in fruit-gatherers
US192585A (en) * 1877-07-03 Improvement in boot and shoe sole trimming machines
US199461A (en) * 1878-01-22 Improvement in fire-escapes
US199538A (en) * 1878-01-22 Improvement in barbed fences
US202720A (en) * 1878-04-23 Improvement in mailing-packages
US214851A (en) * 1879-04-29 Improvement in sheet-metal boxes
US4529624A (en) * 1982-06-28 1985-07-16 Dennison Manufacturing Co. Discoloration resistant heat transfer labeling
US4689338A (en) * 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
US4698348A (en) * 1983-11-18 1987-10-06 Riker Laboratories, Inc. 1H-imidazo[4,5-c]quinolines and their use as bronchodilating agents
US4988815A (en) * 1989-10-26 1991-01-29 Riker Laboratories, Inc. 3-Amino or 3-nitro quinoline compounds which are intermediates in preparing 1H-imidazo[4,5-c]quinolines
US5037986A (en) * 1989-03-23 1991-08-06 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo[4,5-c]quinolin-4-amines
US5175296A (en) * 1991-03-01 1992-12-29 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]quinolin-4-amines and processes for their preparation
US5238944A (en) * 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US5266575A (en) * 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
US5268376A (en) * 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5352784A (en) * 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5367076A (en) * 1990-10-05 1994-11-22 Minnesota Mining And Manufacturing Company Process for imidazo[4,5-C]quinolin-4-amines
US5376501A (en) * 1993-04-27 1994-12-27 Agfa-Gevaert, N.V. Process for incorporation of a water-insoluble substance into a hydrophilic layer
US5389640A (en) * 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5395937A (en) * 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
US5446153A (en) * 1993-07-15 1995-08-29 Minnesota Mining And Manufacturing Company Intermediates for imidazo[4,5-c]pyridin-4-amines
US5482936A (en) * 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
US5693811A (en) * 1996-06-21 1997-12-02 Minnesota Mining And Manufacturing Company Process for preparing tetrahdroimidazoquinolinamines
US5741908A (en) * 1996-06-21 1998-04-21 Minnesota Mining And Manufacturing Company Process for reparing imidazoquinolinamines
US5756747A (en) * 1989-02-27 1998-05-26 Riker Laboratories, Inc. 1H-imidazo 4,5-c!quinolin-4-amines
US5939090A (en) * 1996-12-03 1999-08-17 3M Innovative Properties Company Gel formulations for topical drug delivery
US6028076A (en) * 1996-07-03 2000-02-22 Japan Energy Corporation Purine derivative
US6039969A (en) * 1996-10-25 2000-03-21 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
US6069149A (en) * 1997-01-09 2000-05-30 Terumo Kabushiki Kaisha Amide derivatives and intermediates for the synthesis thereof
US6083505A (en) * 1992-04-16 2000-07-04 3M Innovative Properties Company 1H-imidazo[4,5-C]quinolin-4-amines as vaccine adjuvants
US6110929A (en) * 1998-07-28 2000-08-29 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
US6113918A (en) * 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
US6194425B1 (en) * 1997-12-11 2001-02-27 3M Innovative Properties Company Imidazonaphthyridines
US6194388B1 (en) * 1994-07-15 2001-02-27 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) * 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6245776B1 (en) * 1999-01-08 2001-06-12 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US6303347B1 (en) * 1997-05-08 2001-10-16 Corixa Corporation Aminoalkyl glucosaminide phosphate compounds and their use as adjuvants and immunoeffectors
US6329381B1 (en) * 1997-11-28 2001-12-11 Sumitomo Pharmaceuticals Company, Limited Heterocyclic compounds
US6331539B1 (en) * 1999-06-10 2001-12-18 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6339068B1 (en) * 1997-05-20 2002-01-15 University Of Iowa Research Foundation Vectors and methods for immunization or therapeutic protocols
US6376501B1 (en) * 1997-12-22 2002-04-23 Japan Energy Corporation Type 2 helper T cell-selective immune response suppressors
US6376669B1 (en) * 1999-11-05 2002-04-23 3M Innovative Properties Company Dye labeled imidazoquinoline compounds
US6387938B1 (en) * 1996-07-05 2002-05-14 Mochida Pharmaceutical Co., Ltd. Benzimidazole derivatives
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6426334B1 (en) * 1997-04-30 2002-07-30 Hybridon, Inc. Oligonucleotide mediated specific cytokine induction and reduction of tumor growth in a mammal
US6451810B1 (en) * 1999-06-10 2002-09-17 3M Innovative Properties Company Amide substituted imidazoquinolines
US6476000B1 (en) * 1999-08-13 2002-11-05 Hybridon, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
US6518265B1 (en) * 1998-08-12 2003-02-11 Hokuriku Seiyaku Co., Ltd. 1H-imidazopyridine derivatives
US6525028B1 (en) * 2002-02-04 2003-02-25 Corixa Corporation Immunoeffector compounds
US6525064B1 (en) * 2000-12-08 2003-02-25 3M Innovative Properties Company Sulfonamido substituted imidazopyridines
US6541485B1 (en) * 1999-06-10 2003-04-01 3M Innovative Properties Company Urea substituted imidazoquinolines
US6545016B1 (en) * 2000-12-08 2003-04-08 3M Innovative Properties Company Amide substituted imidazopyridines
US6545017B1 (en) * 2000-12-08 2003-04-08 3M Innovative Properties Company Urea substituted imidazopyridines
US6558951B1 (en) * 1999-02-11 2003-05-06 3M Innovative Properties Company Maturation of dendritic cells with immune response modifying compounds
US6573273B1 (en) * 1999-06-10 2003-06-03 3M Innovative Properties Company Urea substituted imidazoquinolines
US6649172B2 (en) * 2000-03-17 2003-11-18 Corixa Corporation Amphipathic aldehydes and their uses as adjuvants and immunoeffectors
US6656938B2 (en) * 2000-12-08 2003-12-02 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660735B2 (en) * 2000-12-08 2003-12-09 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660747B2 (en) * 2000-12-08 2003-12-09 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6664264B2 (en) * 2000-12-08 2003-12-16 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6664265B2 (en) * 2000-12-08 2003-12-16 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6677348B2 (en) * 2000-12-08 2004-01-13 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
US6677349B1 (en) * 2001-12-21 2004-01-13 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6677347B2 (en) * 2000-12-08 2004-01-13 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
US6706728B2 (en) * 1999-01-08 2004-03-16 3M Innovative Properties Company Systems and methods for treating a mucosal surface
US6743920B2 (en) * 2002-05-29 2004-06-01 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
US6756382B2 (en) * 1999-06-10 2004-06-29 3M Innovative Properties Company Amide substituted imidazoquinolines
US6818650B2 (en) * 2002-09-26 2004-11-16 3M Innovative Properties Company 1H-imidazo dimers
US20050119273A1 (en) * 2003-06-20 2005-06-02 Coley Pharmaceutical Gmbh Small molecule toll-like receptor (TLR) antagonists

Family Cites Families (250)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3314941A (en) 1964-06-23 1967-04-18 American Cyanamid Co Novel substituted pyridodiazepins
DE1645976A1 (en) 1966-06-18 1971-01-21 Ajinomoto Kk Process for the preparation of adenosine and 2 ', 3'-O-isopropylidene adenosine
ZA704419B (en) 1969-07-21 1971-04-28 Ici Australia Ltd Injectable aqueous solutions of tetramisole
US3692907A (en) 1970-10-27 1972-09-19 Richardson Merrell Inc Treating viral infections with bis-basic ethers and thioethers of fluorenone and fluorene and pharmaceutical compositons of the same
US3917624A (en) 1972-09-27 1975-11-04 Pfizer Process for producing 2-amino-nicotinonitrile intermediates
US4006237A (en) 1973-10-11 1977-02-01 Beecham Group Limited Tetrahydrocarbostyril derivatives for the prophylaxis of asthma, hayfever and rhinitis
US3891660A (en) 1974-02-07 1975-06-24 Squibb & Sons Inc Derivatives of 1H-imidazo{8 4,5-c{9 pyridine-7-carboxylic acids and esters
US3899508A (en) 1974-04-12 1975-08-12 Lilly Co Eli 5-(2-Aminophenyl)pyrazole-3-carboxylic acids and esters thereof
DE2423389A1 (en) 1974-05-14 1975-12-04 Hoechst Ag PSYCHOTROPIC MEDICINAL PRODUCTS AND THE METHOD OF MANUFACTURING THEREOF
JPS6016438B2 (en) 1976-10-14 1985-04-25 ウェルファイド株式会社 Imidazoquinoline derivatives
US4381344A (en) 1980-04-25 1983-04-26 Burroughs Wellcome Co. Process for producing deoxyribosides using bacterial phosphorylase
DE3204126A1 (en) 1982-02-06 1983-08-11 Bayer Ag, 5090 Leverkusen PYRAZOLOXAZINE, -THIAZINE, -CHINOLINE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A MEDICINAL PRODUCT
US4758574A (en) 1982-05-03 1988-07-19 Eli Lilly And Company 2-phenylimidazio (4,5-c) pyridines
US4563525A (en) 1983-05-31 1986-01-07 Ici Americas Inc. Process for preparing pyrazolopyridine compounds
CA1271477A (en) 1983-11-18 1990-07-10 John F. Gerster 1h-imidazo[4,5-c]quinolin-4-amines
JPS61112075A (en) 1984-11-05 1986-05-30 Shionogi & Co Ltd Thienylpyrazoloquinoline derivative
US4593821A (en) 1985-04-25 1986-06-10 Laros Equipment Company, Inc. Belt separator for blow molding parts
US4668686A (en) 1985-04-25 1987-05-26 Bristol-Myers Company Imidazoquinoline antithrombrogenic cardiotonic agents
US4698346A (en) 1985-05-08 1987-10-06 Usv Pharmaceutical Corporation Thiazolo[5,4-h]quinoline compounds useful as anti-allergy agents
US4826830A (en) 1985-07-31 1989-05-02 Jui Han Topical application of glyciphosphoramide
CA1306260C (en) 1985-10-18 1992-08-11 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
HU197019B (en) 1985-11-12 1989-02-28 Egyt Gyogyszervegyeszeti Gyar Process for producing thiqzolo (4,5-c) quinoline derivatives and pharmaceuticals comprising same
US4837378A (en) 1986-01-15 1989-06-06 Curatek Pharmaceuticals, Inc. Topical metronidazole formulations and therapeutic uses thereof
JPS6310787A (en) 1986-03-06 1988-01-18 Takeda Chem Ind Ltd Nucleotide analog, production thereof and antiviral agent
US4775674A (en) 1986-05-23 1988-10-04 Bristol-Myers Company Imidazoquinolinylether derivatives useful as phosphodiesterase and blood aggregation inhibitors
CA1287061C (en) 1986-06-27 1991-07-30 Roche Holding Ltd. Pyridine ethanolamine derivatives
US5500228A (en) 1987-05-26 1996-03-19 American Cyanamid Company Phase separation-microencapsulated pharmaceuticals compositions useful for alleviating dental disease
US4880779A (en) 1987-07-31 1989-11-14 Research Corporation Technologies, Inc. Method of prevention or treatment of AIDS by inhibition of human immunodeficiency virus
US4774339A (en) 1987-08-10 1988-09-27 Molecular Probes, Inc. Chemically reactive dipyrrometheneboron difluoride dyes
JPH01180156A (en) 1988-01-12 1989-07-18 Nec Corp Packet switching circuit
US5536743A (en) 1988-01-15 1996-07-16 Curatek Pharmaceuticals Limited Partnership Intravaginal treatment of vaginal infections with buffered metronidazole compositions
US5225183A (en) 1988-12-06 1993-07-06 Riker Laboratories, Inc. Medicinal aerosol formulations
US5736553A (en) 1988-12-15 1998-04-07 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo 4,5-C!quinolin-4-amine
DK0385630T3 (en) 1989-02-27 1997-05-12 Riker Laboratories Inc 1H-imidazo (4,5-c) quinoline-4-amines as antivirals
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
NZ232740A (en) 1989-04-20 1992-06-25 Riker Laboratories Inc Solution for parenteral administration comprising a 1h-imidazo(4,5-c) quinolin-4-amine derivative, an acid and a tonicity adjuster
US5750134A (en) 1989-11-03 1998-05-12 Riker Laboratories, Inc. Bioadhesive composition and patch
EP0507863A4 (en) 1989-12-28 1993-07-07 Virginia Commonwealth University Sigma receptor ligands and the use thereof
US5274113A (en) 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
EP0527818A4 (en) 1990-04-30 1993-09-15 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of arachidonic acid metabolism
AU653524B2 (en) 1990-06-08 1994-10-06 Roussel-Uclaf New imidazole derivatives, their preparation process, the new intermediates obtained, their use as medicaments and the pharmaceutical compositions containing them
JP2660086B2 (en) 1990-07-03 1997-10-08 明治製菓株式会社 Agent for improving brain and cardiac dysfunction
MX9203481A (en) 1990-10-18 1992-07-01 Minnesota Mining & Mfg FORMULATIONS.
US5248782A (en) 1990-12-18 1993-09-28 Molecular Probes, Inc. Long wavelength heteroaryl-substituted dipyrrometheneboron difluoride dyes
JPH04327587A (en) 1991-04-26 1992-11-17 Asahi Chem Ind Co Ltd 6'-c-alkyl-3-deazaneplanocin a derivative, its production and use
US5187288A (en) 1991-05-22 1993-02-16 Molecular Probes, Inc. Ethenyl-substituted dipyrrometheneboron difluoride dyes and their synthesis
TW300219B (en) 1991-09-14 1997-03-11 Hoechst Ag
PH31245A (en) 1991-10-30 1998-06-18 Janssen Pharmaceutica Nv 1,3-Dihydro-2H-imidazoÄ4,5-BÜ-quinolin-2-one derivatives.
US5378848A (en) 1992-02-12 1995-01-03 Shionogi & Co., Ltd. Condensed imidazopyridine derivatives
DK0641192T3 (en) 1992-05-18 1998-03-02 Minnesota Mining & Mfg Transmucosal drug delivery device
US5352680A (en) 1992-07-15 1994-10-04 Regents Of The University Of Minnesota Delta opioid receptor antagonists to block opioid agonist tolerance and dependence
US6608201B2 (en) 1992-08-28 2003-08-19 3M Innovative Properties Company Process for preparing 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
ES2122261T3 (en) 1993-03-17 1998-12-16 Minnesota Mining & Mfg AEROSOL FORMULATION CONTAINING A DISPERSION ADJUVANT DERIVED FROM AN ESTER, AMIDA OR MERCAPTOESTER.
DE4309969A1 (en) 1993-03-26 1994-09-29 Bayer Ag Substituted hetero-fused imidazoles
US5648516A (en) 1994-07-20 1997-07-15 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
CA2131680C (en) 1993-09-17 2006-11-07 Gerhard Stucky Process for preparing imidazopyridine derivatives
US5837809A (en) 1995-08-11 1998-11-17 Oregon Health Sciences University Mammalian opioid receptor ligand and uses
JPH07163368A (en) 1993-12-15 1995-06-27 Hayashibara Biochem Lab Inc Recombinant dna and transformant containing the same recombinant dna
TW574214B (en) 1994-06-08 2004-02-01 Pfizer Corticotropin releasing factor antagonists
US5612377A (en) 1994-08-04 1997-03-18 Minnesota Mining And Manufacturing Company Method of inhibiting leukotriene biosynthesis
US5644063A (en) 1994-09-08 1997-07-01 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]pyridin-4-amine intermediates
GB9420168D0 (en) 1994-10-06 1994-11-23 Boots Co Plc Therapeutic agents
US5571819A (en) 1994-11-22 1996-11-05 Sabb; Annmarie L. Imidazopyridines as muscarinic agents
US6071949A (en) 1995-03-14 2000-06-06 The United States Of America As Represented By The Department Of Health And Human Services Use of lipoxygenase inhibitors as anti-cancer therapeutic and intervention agents
US5585612A (en) 1995-03-20 1996-12-17 Harp Enterprises, Inc. Method and apparatus for voting
FR2732605B1 (en) 1995-04-07 1997-05-16 Pasteur Merieux Serums Vacc COMPOSITION FOR INDUCING MUCOSAL IMMUNE RESPONSE
US5766789A (en) 1995-09-29 1998-06-16 Energetics Systems Corporation Electrical energy devices
DK0778277T3 (en) 1995-12-08 2003-10-27 Pfizer Substituted heterocyclic derivatives as CRF antagonists
JPH09208584A (en) 1996-01-29 1997-08-12 Terumo Corp Amide derivative, pharmaceutical preparation containing the same, and intermediate for synthesizing the same
US5939047A (en) 1996-04-16 1999-08-17 Jernberg; Gary R. Local delivery of chemotherapeutic agents for treatment of periodontal disease
US5861268A (en) 1996-05-23 1999-01-19 Biomide Investment Limited Partnership Method for induction of tumor cell apoptosis with chemical inhibitors targeted to 12-lipoxygenase
US6123957A (en) 1997-07-16 2000-09-26 Jernberg; Gary R. Delivery of agents and method for regeneration of periodontal tissues
US6077349A (en) 1997-08-20 2000-06-20 Sony Corporation Method and apparatus for manufacturing disc-shaped recording medium
US6309623B1 (en) 1997-09-29 2001-10-30 Inhale Therapeutic Systems, Inc. Stabilized preparations for use in metered dose inhalers
US6121323A (en) 1997-12-03 2000-09-19 3M Innovative Properties Company Bishydroxyureas
JPH11222432A (en) 1998-02-03 1999-08-17 Terumo Corp Preparation for external use containing amide derivative inducing interferon
US6114058A (en) 1998-05-26 2000-09-05 Siemens Westinghouse Power Corporation Iron aluminide alloy container for solid oxide fuel cells
US5962636A (en) 1998-08-12 1999-10-05 Amgen Canada Inc. Peptides capable of modulating inflammatory heart disease
US6518280B2 (en) 1998-12-11 2003-02-11 3M Innovative Properties Company Imidazonaphthyridines
US6486168B1 (en) 1999-01-08 2002-11-26 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
EP1495758A3 (en) 1999-01-08 2005-04-13 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US6294271B1 (en) 1999-02-12 2001-09-25 Shin-Etsu Chemical Co., Ltd. Flip-chip type semiconductor device sealing material and flip-chip type semiconductor device
JP2000247884A (en) 1999-03-01 2000-09-12 Sumitomo Pharmaceut Co Ltd Arachidonic acid-induced skin disease-treating agent
WO2000061765A2 (en) 1999-04-12 2000-10-19 Lexicon Genetics Incorporated Lipoxygenase proteins and polynucleotides encoding the same
US6315985B1 (en) 1999-06-18 2001-11-13 3M Innovative Properties Company C-17/21 OH 20-ketosteroid solution aerosol products with enhanced chemical stability
US6432989B1 (en) 1999-08-27 2002-08-13 Pfizer Inc Use of CRF antagonists to treat circadian rhythm disorders
CO5271670A1 (en) 1999-10-29 2003-04-30 Pfizer Prod Inc ANTIGONISTS OF THE CORTICITROPINE RELEASE FACTOR AND RELATED COMPOSITIONS
ATE313318T1 (en) 1999-10-29 2006-01-15 Nektar Therapeutics DRY POWDER COMPOSITIONS WITH IMPROVED DISPERSITY
US6916925B1 (en) 1999-11-05 2005-07-12 3M Innovative Properties Co. Dye labeled imidazoquinoline compounds
US6313156B1 (en) 1999-12-23 2001-11-06 Icos Corporation Thiazole compounds as cyclic-AMP-specific phosphodiesterase inhibitors
US20040023870A1 (en) * 2000-01-21 2004-02-05 Douglas Dedera Methods of therapy and diagnosis using targeting of cells that express toll-like receptor proteins
GB0001704D0 (en) 2000-01-25 2000-03-15 Glaxo Group Ltd Protein
MXPA02007525A (en) 2000-02-09 2002-12-13 Hokuriku Pharmaceutical 1h imidazopyridine derivatives.
US20010046968A1 (en) 2000-03-23 2001-11-29 Zagon Ian S. Opioid growth factor modulates angiogenesis
HUP0300580A3 (en) 2000-03-30 2006-06-28 Shionogi & Co Novel process for producing fused imidazopyridine derivative and novel crystal form
US6894060B2 (en) 2000-03-30 2005-05-17 3M Innovative Properties Company Method for the treatment of dermal lesions caused by envenomation
US20020137101A1 (en) 2000-05-19 2002-09-26 Meyers Rachel A. 46638, a novel human lipoxygenase family member and uses thereof
DE10020465A1 (en) 2000-04-26 2001-11-08 Osram Opto Semiconductors Gmbh Radiation-emitting semiconductor component with luminescence conversion element
DE10029580C1 (en) 2000-06-15 2002-01-10 Ferton Holding Sa Device for removing body stones with an intracorporeal lithotripter
US6387383B1 (en) 2000-08-03 2002-05-14 Dow Pharmaceutical Sciences Topical low-viscosity gel composition
US6900016B1 (en) 2000-09-08 2005-05-31 Applera Corporation Polymorphisms in known genes associated with inflammatory autoimmune disease, methods of detection and uses thereof
US20020055517A1 (en) 2000-09-15 2002-05-09 3M Innovative Properties Company Methods for delaying recurrence of herpes virus symptoms
WO2002022809A2 (en) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
GB0023008D0 (en) 2000-09-20 2000-11-01 Glaxo Group Ltd Improvements in vaccination
JPWO2002046479A1 (en) 2000-12-07 2004-04-08 株式会社青山製作所 Baking treatment method for steel parts
US6667312B2 (en) 2000-12-08 2003-12-23 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6664260B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Heterocyclic ether substituted imidazoquinolines
US20020110840A1 (en) 2000-12-08 2002-08-15 3M Innovative Properties Company Screening method for identifying compounds that selectively induce interferon alpha
UA74593C2 (en) 2000-12-08 2006-01-16 3M Innovative Properties Co Substituted imidazopyridines
US20020107262A1 (en) 2000-12-08 2002-08-08 3M Innovative Properties Company Substituted imidazopyridines
US20020182274A1 (en) 2001-03-21 2002-12-05 Kung-Ming Lu Methods for inhibiting cancer growth, reducing infection and promoting general health with a fermented soy extract
CA2444130C (en) 2001-04-17 2010-12-21 Sumitomo Pharmaceuticals Company, Limited Adenine derivatives
US20020193729A1 (en) 2001-04-20 2002-12-19 Cormier Michel J.N. Microprojection array immunization patch and method
WO2002085933A1 (en) 2001-04-20 2002-10-31 The Institute For Systems Biology Toll-like receptor 5 ligands and methods of use
US6627639B2 (en) 2001-04-26 2003-09-30 Wyeth Uses for indoletetrahydropyridine derivatives of 2,3-dihydro-7H-[1,4]dioxino-[2,3-e]indole
US7226928B2 (en) 2001-06-15 2007-06-05 3M Innovative Properties Company Methods for the treatment of periodontal disease
US6756399B2 (en) 2001-06-29 2004-06-29 The United States Of America As Represented By The Department Of Health And Human Services Use of lipoxygenase inhibitors and PPAR ligands as anti-cancer therapeutic and intervention agents
CA2453664A1 (en) 2001-07-16 2003-01-30 Takayuki Kasai Process for preparation of amidine derivatives
JP2005501550A (en) 2001-08-30 2005-01-20 スリーエム イノベイティブ プロパティズ カンパニー Maturation of plasmacytoid dendritic cells using immune response modifier molecules
ATE511840T1 (en) 2001-10-09 2011-06-15 Amgen Inc IMIDAZOLE DERIVATIVES AS ANTI-INFLAMMATORY AGENTS
US20030139364A1 (en) * 2001-10-12 2003-07-24 University Of Iowa Research Foundation Methods and products for enhancing immune responses using imidazoquinoline compounds
DE60230340D1 (en) 2001-11-16 2009-01-22 3M Innovative Properties Co N-Ä4- (4-amino-2-ethyl-1H-imidazoÄ4,5-quinolin-1-yl) -butyl-methanesulfonamide, pharmaceutical composition containing the same and their use
AU2002364897A1 (en) 2001-11-17 2003-06-10 Maria Martinez-Colon Imiquimod therapies
CN101033242A (en) 2001-11-27 2007-09-12 安那迪斯药品股份有限公司 Antenna system for a level measurement apparatus
NZ532769A (en) 2001-11-29 2005-12-23 3M Innovative Properties Co Pharmaceutical formulations comprising an immune response modifier
US6775514B2 (en) 2002-01-11 2004-08-10 Xerox Corporation Substrate size monitoring system for use in copier/printers
US20050281813A1 (en) 2002-02-14 2005-12-22 Nuvelo, Inc. Methods of therapy and diagnosis using targeting of cells that express toll-like receptor proteins
ES2541132T3 (en) 2002-02-22 2015-07-16 Meda Ab Method to reduce and treat UV-B-induced immunosuppression
US20030185835A1 (en) 2002-03-19 2003-10-02 Braun Ralph P. Adjuvant for vaccines
EP3006043B1 (en) 2002-04-04 2019-05-29 Zoetis Belgium S.A. Immunostimulatory g,u-containing oligoribonucleotides
CN101837003B (en) 2002-04-30 2013-09-04 尤尼根公司 Formulation of a mixture of free-B-ring flavonoids and flavans as a therapeutic agent
GB0211649D0 (en) 2002-05-21 2002-07-03 Novartis Ag Organic compounds
MXPA04012199A (en) 2002-06-07 2005-02-25 3M Innovative Properties Co Ether substituted imidazopyridines.
CN1315828C (en) 2002-07-23 2007-05-16 特瓦药厂私人有限公司 Preparation of 1H-imidazo [4,5-C] quinolin-4-amines via 1H-imidazo [4,5-C] quinolin-4-phthalimide intermediates
JP2005537287A (en) 2002-07-26 2005-12-08 テバ ジョジセルジャール レースベニュタールシャシャーグ 1H-imidazo [4,5-c] quinolin-4-amine with novel 1H-imidazo [4,5-c] quinolin-4-cyano and 1H-imidazo [4,5-c] quinoline-4-carboxamide intermediates Manufacturing
EP1545597B1 (en) 2002-08-15 2010-11-17 3M Innovative Properties Company Immunostimulatory compositions and methods of stimulating an immune response
EP1556053A4 (en) 2002-10-31 2006-04-19 Amgen Inc Antiinflammation agents
WO2004043913A2 (en) 2002-11-08 2004-05-27 Trimeris, Inc. Hetero-substituted benzimidazole compounds and antiviral uses thereof
WO2004053452A2 (en) 2002-12-11 2004-06-24 3M Innovative Properties Company Assays relating to toll-like receptor activity
AU2003287324A1 (en) 2002-12-11 2004-06-30 3M Innovative Properties Company Gene expression systems and recombinant cell lines
WO2004058759A1 (en) 2002-12-20 2004-07-15 3M Innovative Properties Company Aryl / hetaryl substituted imidazoquinolines
CA2511538C (en) 2002-12-30 2013-11-26 3M Innovative Properties Company Immunostimulatory combinations
US7375180B2 (en) 2003-02-13 2008-05-20 3M Innovative Properties Company Methods and compositions related to IRM compounds and Toll-like receptor 8
EP1599726A4 (en) 2003-02-27 2009-07-22 3M Innovative Properties Co Selective modulation of tlr-mediated biological activity
CA2517528A1 (en) 2003-03-04 2004-09-16 3M Innovative Properties Company Prophylactic treatment of uv-induced epidermal neoplasia
US7163947B2 (en) 2003-03-07 2007-01-16 3M Innovative Properties Company 1-Amino 1H-imidazoquinolines
EP1605943A4 (en) 2003-03-07 2008-01-16 3M Innovative Properties Co 1-amino 1h-imidazoquinolines
AU2004220465A1 (en) 2003-03-13 2004-09-23 3M Innovative Properties Company Method of tattoo removal
CA2518282C (en) 2003-03-13 2012-11-06 3M Innovative Properties Company Methods of improving skin quality
US7699057B2 (en) 2003-03-13 2010-04-20 3M Innovative Properties Company Methods for treating skin lesions
US20040192585A1 (en) 2003-03-25 2004-09-30 3M Innovative Properties Company Treatment for basal cell carcinoma
WO2004087049A2 (en) 2003-03-25 2004-10-14 3M Innovative Properties Company Selective activation of cellular activities mediated through a common toll-like receptor
US7893096B2 (en) 2003-03-28 2011-02-22 Novartis Vaccines And Diagnostics, Inc. Use of small molecule compounds for immunopotentiation
US20040265351A1 (en) 2003-04-10 2004-12-30 Miller Richard L. Methods and compositions for enhancing immune response
US20040202720A1 (en) 2003-04-10 2004-10-14 3M Innovative Properties Company Delivery of immune response modifier compounds using metal-containing particulate support materials
US20040214851A1 (en) 2003-04-28 2004-10-28 3M Innovative Properties Company Compositions and methods for induction of opioid receptors
WO2004110991A2 (en) 2003-06-06 2004-12-23 3M Innovative Properties Company PROCESS FOR IMIDAZO[4,5-c]PYRIDIN-4-AMINES
AR044466A1 (en) 2003-06-06 2005-09-14 3M Innovative Properties Co PROCESS FOR THE PREPARATION OF IMIDAZO [4,5-C] PIRIDIN-4-AMINAS
MY157827A (en) 2003-06-27 2016-07-29 3M Innovative Properties Co Sulfonamide substituted imidazoquinolines
US20050106300A1 (en) 2003-06-30 2005-05-19 Purdue Research Foundation Method for producing a material having an increased solubility in alcohol
CA2534313C (en) 2003-08-05 2013-03-19 3M Innovative Properties Company Formulations containing an immune response modifier
TW200510412A (en) 2003-08-12 2005-03-16 3M Innovative Properties Co Oxime substituted imidazo-containing compounds
CA2535338C (en) 2003-08-14 2013-05-28 3M Innovative Properties Company Substituted 1h-imidazo[4,5-c]pyridin-4-amines,1h-imidazo[4,5-c]quinolin -4-amines and 1h-imidazo[4,5-c]naphthyridin-4-amines as immune response modifiers
JP2007504145A (en) 2003-08-25 2007-03-01 スリーエム イノベイティブ プロパティズ カンパニー Immunostimulatory combinations and treatments
CA2536136C (en) 2003-08-27 2012-10-30 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
JP2007504269A (en) 2003-09-05 2007-03-01 スリーエム イノベイティブ プロパティズ カンパニー Method for treating CD5 + B cell lymphoma
NZ545536A (en) 2003-09-05 2010-04-30 Anadys Pharmaceuticals Inc TLR7 ligands for the treatment of hepatitis C
GB0321615D0 (en) 2003-09-15 2003-10-15 Glaxo Group Ltd Improvements in vaccination
EP1664342A4 (en) 2003-09-17 2007-12-26 3M Innovative Properties Co Selective modulation of tlr gene expression
WO2005033049A2 (en) 2003-10-01 2005-04-14 Taro Pharmaceuticals U.S.A., Inc. METHOD OF PREPARING 4-AMINO-1H-IMIDAZO(4,5-c)QUINOLINES AND ACID ADDITION SALTS THEREOF
NZ546274A (en) 2003-10-03 2009-12-24 3M Innovative Properties Co Pyrazolopyridines and analags thereof
US20090075980A1 (en) 2003-10-03 2009-03-19 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and Analogs Thereof
JP5043435B2 (en) 2003-10-03 2012-10-10 スリーエム イノベイティブ プロパティズ カンパニー Alkoxy substituted imidazoquinolines
US7544697B2 (en) 2003-10-03 2009-06-09 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and analogs thereof
AU2004285575A1 (en) 2003-10-31 2005-05-12 3M Innovative Properties Company Neutrophil activation by immune response modifier compounds
US20050239733A1 (en) 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
ITMI20032121A1 (en) 2003-11-04 2005-05-05 Dinamite Dipharma Spa In Forma Abbr Eviata Dipharm PROCEDURE FOR THE PREPARATION OF IMIQUIMOD AND ITS INTERMEDIATES
AU2004291122A1 (en) 2003-11-14 2005-06-02 3M Innovative Properties Company Hydroxylamine substituted imidazo ring compounds
CN1906193A (en) 2003-11-14 2007-01-31 3M创新有限公司 Oxime substituted imidazo ring compounds
CU23404A1 (en) 2003-11-19 2009-08-04 Ct Ingenieria Genetica Biotech NEISSERIA MENINGITIDIS CAPSULAR POLYSACARIDS AS MUCOSOPT IMMUNOPOTENTIZERS AND RESULTING FORMULATIONS
CN1902200A (en) 2003-11-21 2007-01-24 诺瓦提斯公司 1h-imidazoquinoline derivatives as protein kinase inhibitors
AR046845A1 (en) 2003-11-21 2005-12-28 Novartis Ag DERIVATIVES OF 1H-IMIDAZO [4,5-C] QUINOLINE FOR THE TREATMENT OF PROTEIN-KINASE DEPENDENT DISEASES
AR046781A1 (en) 2003-11-25 2005-12-21 3M Innovative Properties Co IMIDAZOQUINOLINE DERIVATIVES. PHARMACEUTICAL COMPOSITIONS.
EP1686992A4 (en) 2003-11-25 2009-11-04 3M Innovative Properties Co Hydroxylamine and oxime substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
US20050226878A1 (en) 2003-12-02 2005-10-13 3M Innovative Properties Company Therapeutic combinations and methods including IRM compounds
EP1689361A4 (en) 2003-12-02 2009-06-17 3M Innovative Properties Co Therapeutic combinations and methods including irm compounds
WO2005076783A2 (en) 2003-12-04 2005-08-25 3M Innovative Properties Company Sulfone substituted imidazo ring ethers
WO2005066170A1 (en) 2003-12-29 2005-07-21 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
CA2552101A1 (en) 2003-12-29 2005-07-21 3M Innovative Properties Company Piperazine, [1,4]diazepane, [1,4]diazocane, and [1,5]diazocane fused imidazo ring compounds
WO2005065678A1 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Immunomodulatory combinations
US20050239735A1 (en) 2003-12-30 2005-10-27 3M Innovative Properties Company Enhancement of immune responses
WO2005066169A2 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Imidazoquinolinyl, imidazopyridinyl, and imidazonaphthyridinyl sulfonamides
WO2005094531A2 (en) 2004-03-24 2005-10-13 3M Innovative Properties Company Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
EP1735010A4 (en) 2004-04-09 2008-08-27 3M Innovative Properties Co Methods, compositions, and preparations for delivery of immune response modifiers
US20050267145A1 (en) 2004-05-28 2005-12-01 Merrill Bryon A Treatment for lung cancer
US20080015184A1 (en) 2004-06-14 2008-01-17 3M Innovative Properties Company Urea Substituted Imidazopyridines, Imidazoquinolines, and Imidazonaphthyridines
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
WO2006009832A1 (en) 2004-06-18 2006-01-26 3M Innovative Properties Company Substituted imidazo ring systems and methods
US7915281B2 (en) 2004-06-18 2011-03-29 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and method
EP1765348B1 (en) 2004-06-18 2016-08-03 3M Innovative Properties Company Substituted imidazoquinolines, imidazopyridines, and imidazonaphthyridines
WO2006093514A2 (en) 2004-06-18 2006-09-08 3M Innovative Properties Company Aryl and arylalkylenyl substituted thiazoloquinolines and thiazolonaphthyridines
US7897609B2 (en) 2004-06-18 2011-03-01 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
WO2006009826A1 (en) 2004-06-18 2006-01-26 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US20090270443A1 (en) 2004-09-02 2009-10-29 Doris Stoermer 1-amino imidazo-containing compounds and methods
AU2005282523A1 (en) 2004-09-02 2006-03-16 3M Innovative Properties Company 2-amino 1H imidazo ring systems and methods
AU2005282726B2 (en) 2004-09-02 2011-06-02 3M Innovative Properties Company 1-alkoxy 1H-imidazo ring systems and methods
WO2006028451A1 (en) 2004-09-03 2006-03-16 3M Innovative Properties Company 1-amino 1-h-imidazoquinolines
US20080213308A1 (en) 2004-09-14 2008-09-04 Nicholas Valiante Imidazoquinoline Compounds
US20100113565A1 (en) 2004-12-08 2010-05-06 Gorden Keith B Immunostimulatory combinations and methods
WO2006063072A2 (en) 2004-12-08 2006-06-15 3M Innovative Properties Company Immunomodulatory compositions, combinations and methods
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
EP1830880A4 (en) 2004-12-30 2008-03-26 Coley Pharm Group Inc Multi-route administration of immune response modifier compounds
WO2006074003A2 (en) 2004-12-30 2006-07-13 3M Innovative Properties Company CHIRAL FUSED [1,2]IMIDAZO[4,5-c] RING COMPOUNDS
WO2006084251A2 (en) 2005-02-04 2006-08-10 Coley Pharmaceutical Group, Inc. Aqueous gel formulations containing immune reponse modifiers
AU2006212765B2 (en) 2005-02-09 2012-02-02 3M Innovative Properties Company Alkyloxy substituted thiazoloquinolines and thiazolonaphthyridines
AU2006338521A1 (en) 2005-02-09 2007-10-11 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted thiazolo(4,5-c) ring compounds and methods
CA2597446A1 (en) 2005-02-11 2006-08-31 Coley Pharmaceutical Group, Inc. Substituted imidazoquinolines and imidazonaphthyridines
JP2008530113A (en) 2005-02-11 2008-08-07 コーリー ファーマシューティカル グループ,インコーポレイテッド Oxime and hydroxyramine substituted imidazo [4,5-c] ring compounds and methods
AU2006213745A1 (en) 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Substituted fused [1,2]imidazo[4,5-c] ring compounds and methods
AU2006216686A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Method of preferentially inducing the biosynthesis of interferon
AU2006216799A1 (en) 2005-02-23 2006-08-31 Coley Pharmaceutical Group, Inc. Hydroxyalkyl substituted imidazonaphthyridines
US8178677B2 (en) 2005-02-23 2012-05-15 3M Innovative Properties Company Hydroxyalkyl substituted imidazoquinolines
JP2008531567A (en) 2005-02-23 2008-08-14 コーリー ファーマシューティカル グループ,インコーポレイテッド Hydroxyalkyl-substituted imidazoquinoline compounds and methods
US7943636B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
CA2602853A1 (en) 2005-04-01 2006-11-16 Coley Pharmaceutical Group, Inc. Ring closing and related methods and intermediates
WO2006107771A2 (en) 2005-04-01 2006-10-12 Coley Pharmaceutical Group, Inc. PYRAZOLO[3,4-c]QUINOLINES, PYRAZOLO[3,4-c]NAPHTHYRIDINES, ANALOGS THEREOF, AND METHODS
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
RS20080128A (en) 2005-09-02 2009-05-06 Pfizer Inc., Hydroxy substituted 1h-imidazopyridines and methods
EA200800782A1 (en) 2005-09-09 2008-08-29 Коли Фармасьютикал Груп, Инк. AMIDA AND CARBAMATE DERIVATIVES N- {2- [4-AMINO-2- (ETOXIMETHYL) -1H-IMIDAZOLO [4,5-c] QUINOLIN-1-IL] -1,1-DIMETHYLETHYL} METHANE SULFONAMIDE AND METHODS
ZA200803029B (en) 2005-09-09 2009-02-25 Coley Pharm Group Inc Amide and carbamate derivatives of alkyl substituted /V-[4-(4-amino-1H-imidazo[4,5-c] quinolin-1-yl)butyl] methane-sulfonamides and methods
AU2006292119A1 (en) 2005-09-23 2007-03-29 3M Innovative Properties Company Method for 1H-imidazo[4,5-c]pyridines and analogs thereof
JP5247458B2 (en) 2005-11-04 2013-07-24 スリーエム・イノベイティブ・プロパティーズ・カンパニー Hydroxy and alkoxy substituted 1H-imidazoquinolines and methods
WO2007062043A1 (en) 2005-11-23 2007-05-31 Coley Pharmaceutical Group Inc. Method of activating murine toll-like receptor 8
MX2008007864A (en) 2005-12-16 2009-03-04 Coley Pharm Group Inc Substituted imidazoquinolines, imidazonaphthyridines, and imidazopyridines, compositions, and methods.
WO2007079171A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for hodgkin's lymphoma
WO2007079169A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for acute myeloid leukemia
EP1968582A4 (en) 2005-12-28 2011-02-16 3M Innovative Properties Co Treatment for cutaneous t cell lymphoma
WO2007079146A1 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc Treatment for non-hodgkin's lymphoma
WO2007079086A1 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Pyrazoloalkyl substituted imidazo ring compounds and methods
WO2007079202A2 (en) 2005-12-28 2007-07-12 Coley Pharmaceutical Group, Inc. Treatment for acute lymhoblastic leukemia
US20090306388A1 (en) 2006-02-10 2009-12-10 Pfizer Inc. Method for substituted ih-imidazo[4,5-c] pyridines
WO2007106854A2 (en) 2006-03-15 2007-09-20 Coley Pharmaceutical Group, Inc. Hydroxy and alkoxy substituted 1h-imidazonaphthyridines and methods
US8088788B2 (en) 2006-03-15 2012-01-03 3M Innovative Properties Company Substituted fused[1,2] imidazo[4,5-c] ring compounds and methods
WO2007143526A2 (en) 2006-06-05 2007-12-13 Coley Pharmaceutical Group, Inc. Substituted tetrahydroimidazonaphthyridines and methods
US7906506B2 (en) 2006-07-12 2011-03-15 3M Innovative Properties Company Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US8178539B2 (en) 2006-09-06 2012-05-15 3M Innovative Properties Company Substituted 3,4,6,7-tetrahydro-5H-1,2a,4a,8-tetraazacyclopenta[cd]phenalenes and methods
WO2008036312A1 (en) 2006-09-19 2008-03-27 Coley Pharmaceutical Group, Inc. Fungicidal methods using immune response modifier compounds
WO2008045543A1 (en) 2006-10-13 2008-04-17 Coley Pharmaceutical Group, Inc. Substituted 4h-imidazo [4, 5, 1-ij] [1, 6] naphthyridine-9-amines and their pharmaceutical use

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US180919A (en) * 1876-08-08 Improvement in clothes-sticks
US214851A (en) * 1879-04-29 Improvement in sheet-metal boxes
US22302A (en) * 1858-12-14 Machine for turning irregular forms
US23870A (en) * 1859-05-03 Island
US55517A (en) * 1866-06-12 Improvement in hand corn - planters
US91491A (en) * 1869-06-15 Improved ash-sifter
US175336A (en) * 1876-03-28 Improvement in grand-piano-forte frames
US130299A (en) * 1872-08-06 Improvement in fruit-baskets
US132079A (en) * 1872-10-08 Improvement in nut-locks
US133913A (en) * 1872-12-10 Improvement in car-brakes
US139364A (en) * 1873-05-27 Improvement in carriage-protectors
US141950A (en) * 1873-08-19 Improvement in stoppers for bottles
US144283A (en) * 1873-11-04 Improvement in match-boxes
US147543A (en) * 1874-02-17 Improvement in piston-packings
US161797A (en) * 1875-04-06 Improvement in combined water-coolers and refrigerators
US162309A (en) * 1875-04-20 Improvement in gas-retorts
US162332A (en) * 1875-04-20 Improvement in machines for coating fabrics
US10007A (en) * 1853-09-13 Gear op variable cut-ofp valves for steau-ehgietes
US110840A (en) * 1871-01-10 Improvement in shoes
US176367A (en) * 1876-04-18 Improvement in safety-locks for fire-arms
US171086A (en) * 1875-12-14 Improvement in throttle-valves
US181130A (en) * 1876-08-15 Improvement in butter-packages
US181211A (en) * 1876-08-15 Improvement in cotton chopper and scraper
US191833A (en) * 1877-06-12 Improvement in fruit-gatherers
US192585A (en) * 1877-07-03 Improvement in boot and shoe sole trimming machines
US199461A (en) * 1878-01-22 Improvement in fire-escapes
US199538A (en) * 1878-01-22 Improvement in barbed fences
US202720A (en) * 1878-04-23 Improvement in mailing-packages
US14779A (en) * 1856-04-29 Xwrench
US4529624A (en) * 1982-06-28 1985-07-16 Dennison Manufacturing Co. Discoloration resistant heat transfer labeling
US4689338A (en) * 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
US4698348A (en) * 1983-11-18 1987-10-06 Riker Laboratories, Inc. 1H-imidazo[4,5-c]quinolines and their use as bronchodilating agents
US5238944A (en) * 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US5756747A (en) * 1989-02-27 1998-05-26 Riker Laboratories, Inc. 1H-imidazo 4,5-c!quinolin-4-amines
US5037986A (en) * 1989-03-23 1991-08-06 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo[4,5-c]quinolin-4-amines
US4988815A (en) * 1989-10-26 1991-01-29 Riker Laboratories, Inc. 3-Amino or 3-nitro quinoline compounds which are intermediates in preparing 1H-imidazo[4,5-c]quinolines
US5367076A (en) * 1990-10-05 1994-11-22 Minnesota Mining And Manufacturing Company Process for imidazo[4,5-C]quinolin-4-amines
US5175296A (en) * 1991-03-01 1992-12-29 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]quinolin-4-amines and processes for their preparation
US5605899A (en) * 1991-03-01 1997-02-25 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5389640A (en) * 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5525612A (en) * 1991-09-04 1996-06-11 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo-[4,5-c]quinolin-4-amines
US5346905A (en) * 1991-09-04 1994-09-13 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo-[4,5-C]quinolin-4-amines
US5268376A (en) * 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5266575A (en) * 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
US6083505A (en) * 1992-04-16 2000-07-04 3M Innovative Properties Company 1H-imidazo[4,5-C]quinolin-4-amines as vaccine adjuvants
US5395937A (en) * 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
US5376501A (en) * 1993-04-27 1994-12-27 Agfa-Gevaert, N.V. Process for incorporation of a water-insoluble substance into a hydrophilic layer
US5352784A (en) * 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5446153A (en) * 1993-07-15 1995-08-29 Minnesota Mining And Manufacturing Company Intermediates for imidazo[4,5-c]pyridin-4-amines
US5494916A (en) * 1993-07-15 1996-02-27 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]pyridin-4-amines
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) * 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6194388B1 (en) * 1994-07-15 2001-02-27 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US5482936A (en) * 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
US5741908A (en) * 1996-06-21 1998-04-21 Minnesota Mining And Manufacturing Company Process for reparing imidazoquinolinamines
US5693811A (en) * 1996-06-21 1997-12-02 Minnesota Mining And Manufacturing Company Process for preparing tetrahdroimidazoquinolinamines
US6028076A (en) * 1996-07-03 2000-02-22 Japan Energy Corporation Purine derivative
US6387938B1 (en) * 1996-07-05 2002-05-14 Mochida Pharmaceutical Co., Ltd. Benzimidazole derivatives
US6200592B1 (en) * 1996-10-25 2001-03-13 3M Innovative Properties Company Immine response modifier compounds for treatment of TH2 mediated and related diseases
US6039969A (en) * 1996-10-25 2000-03-21 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
US5939090A (en) * 1996-12-03 1999-08-17 3M Innovative Properties Company Gel formulations for topical drug delivery
US6069149A (en) * 1997-01-09 2000-05-30 Terumo Kabushiki Kaisha Amide derivatives and intermediates for the synthesis thereof
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6426334B1 (en) * 1997-04-30 2002-07-30 Hybridon, Inc. Oligonucleotide mediated specific cytokine induction and reduction of tumor growth in a mammal
US6113918A (en) * 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
US6303347B1 (en) * 1997-05-08 2001-10-16 Corixa Corporation Aminoalkyl glucosaminide phosphate compounds and their use as adjuvants and immunoeffectors
US6339068B1 (en) * 1997-05-20 2002-01-15 University Of Iowa Research Foundation Vectors and methods for immunization or therapeutic protocols
US6329381B1 (en) * 1997-11-28 2001-12-11 Sumitomo Pharmaceuticals Company, Limited Heterocyclic compounds
US6194425B1 (en) * 1997-12-11 2001-02-27 3M Innovative Properties Company Imidazonaphthyridines
US6376501B1 (en) * 1997-12-22 2002-04-23 Japan Energy Corporation Type 2 helper T cell-selective immune response suppressors
US6110929A (en) * 1998-07-28 2000-08-29 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
US6518265B1 (en) * 1998-08-12 2003-02-11 Hokuriku Seiyaku Co., Ltd. 1H-imidazopyridine derivatives
US6706728B2 (en) * 1999-01-08 2004-03-16 3M Innovative Properties Company Systems and methods for treating a mucosal surface
US6245776B1 (en) * 1999-01-08 2001-06-12 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US6558951B1 (en) * 1999-02-11 2003-05-06 3M Innovative Properties Company Maturation of dendritic cells with immune response modifying compounds
US6573273B1 (en) * 1999-06-10 2003-06-03 3M Innovative Properties Company Urea substituted imidazoquinolines
US6756382B2 (en) * 1999-06-10 2004-06-29 3M Innovative Properties Company Amide substituted imidazoquinolines
US6541485B1 (en) * 1999-06-10 2003-04-01 3M Innovative Properties Company Urea substituted imidazoquinolines
US6451810B1 (en) * 1999-06-10 2002-09-17 3M Innovative Properties Company Amide substituted imidazoquinolines
US6331539B1 (en) * 1999-06-10 2001-12-18 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6476000B1 (en) * 1999-08-13 2002-11-05 Hybridon, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
US6376669B1 (en) * 1999-11-05 2002-04-23 3M Innovative Properties Company Dye labeled imidazoquinoline compounds
US6649172B2 (en) * 2000-03-17 2003-11-18 Corixa Corporation Amphipathic aldehydes and their uses as adjuvants and immunoeffectors
US6683088B2 (en) * 2000-12-08 2004-01-27 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
US6545017B1 (en) * 2000-12-08 2003-04-08 3M Innovative Properties Company Urea substituted imidazopyridines
US6660735B2 (en) * 2000-12-08 2003-12-09 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6656938B2 (en) * 2000-12-08 2003-12-02 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660747B2 (en) * 2000-12-08 2003-12-09 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6664264B2 (en) * 2000-12-08 2003-12-16 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6664265B2 (en) * 2000-12-08 2003-12-16 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6677348B2 (en) * 2000-12-08 2004-01-13 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
US6545016B1 (en) * 2000-12-08 2003-04-08 3M Innovative Properties Company Amide substituted imidazopyridines
US6677347B2 (en) * 2000-12-08 2004-01-13 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
US6525064B1 (en) * 2000-12-08 2003-02-25 3M Innovative Properties Company Sulfonamido substituted imidazopyridines
US6677349B1 (en) * 2001-12-21 2004-01-13 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6525028B1 (en) * 2002-02-04 2003-02-25 Corixa Corporation Immunoeffector compounds
US6743920B2 (en) * 2002-05-29 2004-06-01 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
US6818650B2 (en) * 2002-09-26 2004-11-16 3M Innovative Properties Company 1H-imidazo dimers
US20050119273A1 (en) * 2003-06-20 2005-06-02 Coley Pharmaceutical Gmbh Small molecule toll-like receptor (TLR) antagonists

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080306252A1 (en) * 2000-12-08 2008-12-11 Coley Pharmaceutical Group, Inc. Sulfonamido ether substituted imidazoquinolines
US20040214851A1 (en) * 2003-04-28 2004-10-28 3M Innovative Properties Company Compositions and methods for induction of opioid receptors
US20070066639A1 (en) * 2003-08-12 2007-03-22 Kshirsagar Tushar A Oxime substituted imidazoquinolines
US8673932B2 (en) 2003-08-12 2014-03-18 3M Innovative Properties Company Oxime substituted imidazo-containing compounds
US20090018122A1 (en) * 2003-08-27 2009-01-15 Lindstrom Kyle J Aryloxy and Arylalkyleneoxy Substituted Imidazoquinolines
US7897597B2 (en) 2003-08-27 2011-03-01 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted imidazoquinolines
US20090017076A1 (en) * 2003-09-05 2009-01-15 Coley Pharmaceutical Group, Inc. Treatment for cd5+ b cell lymphoma
US7923429B2 (en) 2003-09-05 2011-04-12 3M Innovative Properties Company Treatment for CD5+ B cell lymphoma
US8871782B2 (en) 2003-10-03 2014-10-28 3M Innovative Properties Company Alkoxy substituted imidazoquinolines
US20090318435A1 (en) * 2003-10-03 2009-12-24 Hays David S Pyrazolopyridines and analogs thereof
US20090075980A1 (en) * 2003-10-03 2009-03-19 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and Analogs Thereof
US7879849B2 (en) 2003-10-03 2011-02-01 3M Innovative Properties Company Pyrazolopyridines and analogs thereof
US20070060754A1 (en) * 2003-10-03 2007-03-15 Lindstrom Kyle J Alkoxy substituted imidazoquinolines
US7897767B2 (en) 2003-11-14 2011-03-01 3M Innovative Properties Company Oxime substituted imidazoquinolines
US20090105295A1 (en) * 2003-11-14 2009-04-23 Coley Pharmaceutical Group, Inc. Hydroxylamine substituted imidazoquinolines
US8598192B2 (en) 2003-11-14 2013-12-03 3M Innovative Properties Company Hydroxylamine substituted imidazoquinolines
US20070072893A1 (en) * 2003-11-25 2007-03-29 Krepski Larry R Substituted imidazo ring systems and methods
US8691837B2 (en) 2003-11-25 2014-04-08 3M Innovative Properties Company Substituted imidazo ring systems and methods
US20090030030A1 (en) * 2003-12-29 2009-01-29 Bonk Jason D Arylalkenyl and arylalkynyl substituted imidazoquinolines
US8802853B2 (en) 2003-12-29 2014-08-12 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
US20090062272A1 (en) * 2003-12-30 2009-03-05 Bonk Jason D Imidazoquinolinyl sulfonamides
US8735421B2 (en) 2003-12-30 2014-05-27 3M Innovative Properties Company Imidazoquinolinyl sulfonamides
US20070219196A1 (en) * 2004-03-24 2007-09-20 Krepski Larry R Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US8697873B2 (en) 2004-03-24 2014-04-15 3M Innovative Properties Company Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
US20070213356A1 (en) * 2004-06-15 2007-09-13 Merrill Bryon A Nitrogen-Containing Heterocyclyl Substituted Imidazoquinolines and Imidazonaphthyridines
US20070287725A1 (en) * 2004-06-18 2007-12-13 3M Innovative Properties Company Isoxazole, Dihydroisoxazole, And Oxadiazole Substituted Imidazo Ring Compounds And Method
US20070219228A1 (en) * 2004-06-18 2007-09-20 Shri Niwas Aryl substituted imidazonaphthyridines
US20070208052A1 (en) * 2004-06-18 2007-09-06 Prince Ryan B Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US7915281B2 (en) 2004-06-18 2011-03-29 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and method
US7897609B2 (en) 2004-06-18 2011-03-01 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US20090270443A1 (en) * 2004-09-02 2009-10-29 Doris Stoermer 1-amino imidazo-containing compounds and methods
US20110070575A1 (en) * 2004-12-08 2011-03-24 Coley Pharmaceutical Group, Inc. Immunomodulatory Compositions, Combinations and Methods
US7943609B2 (en) 2004-12-30 2011-05-17 3M Innovative Proprerties Company Chiral fused [1,2]imidazo[4,5-C] ring compounds
US20080269192A1 (en) * 2004-12-30 2008-10-30 Coley Pharmaceutical Group, Inc. Chiral Fused [1,2]Imidazo[4,5-C] Ring Compounds
US8034938B2 (en) 2004-12-30 2011-10-11 3M Innovative Properties Company Substituted chiral fused [1,2]imidazo[4,5-c] ring compounds
US9603917B2 (en) 2005-01-28 2017-03-28 Galenbio, Inc. Immunologically active compositions
US20110117204A1 (en) * 2005-01-28 2011-05-19 Galenbio, Inc. Immunologically active compositions
US9138467B2 (en) 2005-01-28 2015-09-22 Stipkovits, Laszlo, Dr. Immunologically active compositions
US10071156B2 (en) 2005-02-04 2018-09-11 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
US20090163532A1 (en) * 2005-02-04 2009-06-25 Coley Pharmaceutical Group, Inc. Aqueous Gel Formulations Containing Immune Response Modifiers
US9248127B2 (en) 2005-02-04 2016-02-02 3M Innovative Properties Company Aqueous gel formulations containing immune response modifiers
US7968563B2 (en) 2005-02-11 2011-06-28 3M Innovative Properties Company Oxime and hydroxylamine substituted imidazo[4,5-c] ring compounds and methods
US7943636B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company 1-substituted pyrazolo (3,4-C) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
US7943610B2 (en) 2005-04-01 2011-05-17 3M Innovative Properties Company Pyrazolopyridine-1,4-diamines and analogs thereof
US20080193474A1 (en) * 2005-04-25 2008-08-14 Griesgraber George W Immunostimulatory Compositions
CN101460178A (en) * 2006-04-07 2009-06-17 艾德拉药物股份有限公司 Stabilized immune modulatory RNA (SIMRA) compounds for TLR7 and TLR8
US7906506B2 (en) 2006-07-12 2011-03-15 3M Innovative Properties Company Substituted chiral fused [1,2] imidazo [4,5-c] ring compounds and methods
US20080070907A1 (en) * 2006-07-12 2008-03-20 Coley Pharmaceutical Group, Inc. Substituted chiral fused [1,2] imidazo [4,5-C] ring compounds and methods

Also Published As

Publication number Publication date
US7923429B2 (en) 2011-04-12
AU2004270201A1 (en) 2005-03-17
EP1660026A4 (en) 2008-07-16
JP2007504269A (en) 2007-03-01
WO2005023190A3 (en) 2006-06-01
CA2537763A1 (en) 2005-03-17
US20090017076A1 (en) 2009-01-15
WO2005023190A2 (en) 2005-03-17
EP1660026A2 (en) 2006-05-31

Similar Documents

Publication Publication Date Title
US7923429B2 (en) Treatment for CD5+ B cell lymphoma
Hanna et al. Inhibition of Hedgehog signaling reprograms the dysfunctional immune microenvironment in breast cancer
Dudek et al. First in human phase I trial of 852A, a novel systemic toll-like receptor 7 agonist, to activate innate immune responses in patients with advanced cancer
Zanoni et al. IL-15 cis presentation is required for optimal NK cell activation in lipopolysaccharide-mediated inflammatory conditions
Zhang et al. Mobilization of dendritic cell precursors into the circulation by administration of MIP-1α in mice
Smits et al. The Toll-like receptor 7/8 agonist resiquimod greatly increases the immunostimulatory capacity of human acute myeloid leukemia cells
US20220370606A1 (en) Combination Treatments Of Cancer Comprising A TLR Agonist
Walshaw et al. Toll-like receptor agonists and radiation therapy combinations: an untapped opportunity to induce anticancer immunity and improve tumor control
Chen et al. FLT3 ligand administration after hematopoietic cell transplantation increases circulating dendritic cell precursors that can be activated by CpG oligodeoxynucleotides to enhance T-cell and natural killer cell function
AU2006306521B2 (en) The induction of indoleamine 2,3-dioxygenase in dendritic cells by TLR ligands and uses thereof
Raïch-Regué et al. Intratumoral delivery of mTORC2-deficient dendritic cells inhibits B16 melanoma growth by promoting CD8+ effector T cell responses
AU2023208190A1 (en) Methods relating to activated dendritic cell compositions and immunotherapeutic treatments for subjects with advanced cancers
KR20150092122A (en) Allogeneic Autophagosome-enriched composition for the treatment of disease
Calzado-Villarreal et al. Primary cutaneous CD30+ lymphoproliferative disorders
WO2017201200A1 (en) Therapeutic compositions containing rar-alpha antagonists
EP3331538B1 (en) Modulating inflammasome activation of myeloid-derived suppressor cells for treating gvhd or tumor
KR20230008197A (en) Methods, therapies and uses for treating cancer
CN1863531A (en) Treatment for cd5+ b cell lymphoma
Sandhu Investigating the Anti-Tumoral Mechanism of an Immature Dendritic Cell Targeting Vaccine in Combination with 5-Aza-2’deoxycytidine and IFNα in the B16F10 Murine Melanoma Model
Ma et al. Absence of Stat1 in donor CD4+ T cells promotes the expansion of Tregs and reduces graft-versus-host disease in mice
Lieberman et al. Endogenous CD28 drives CAR T cell responses in multiple myeloma
KR20240009976A (en) How to Generate Improved Immune Cell Populations
Wurzenberger Dendritic cell vaccines in tumor immunotherapy: Immune activation strategies with ligands for the Toll-like receptors 7 and 9
Hammond et al. Sensitization of IL-2 Signaling through
Wurzenberger Dendritic cell vaccines in tumor immunotherapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: 3M INNOVATIVE PROPERTIES COMPANY, MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MILLER, RICHARD L.;SPANER, DAVID E.;REEL/FRAME:015774/0689;SIGNING DATES FROM 20040901 TO 20040903

AS Assignment

Owner name: COLEY PHARMACEUTICAL GROUP, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:3M COMPANY; 3M INNOVATIVE PROPERTIES COMPANY;REEL/FRAME:019945/0698

Effective date: 20070723

STCB Information on status: application discontinuation

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