EP1765313A2 - Verbindungen als immunstimulatoren - Google Patents

Verbindungen als immunstimulatoren

Info

Publication number
EP1765313A2
EP1765313A2 EP05791104A EP05791104A EP1765313A2 EP 1765313 A2 EP1765313 A2 EP 1765313A2 EP 05791104 A EP05791104 A EP 05791104A EP 05791104 A EP05791104 A EP 05791104A EP 1765313 A2 EP1765313 A2 EP 1765313A2
Authority
EP
European Patent Office
Prior art keywords
alkyl
antigens
compound
aryl
heterocyclyl
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.)
Withdrawn
Application number
EP05791104A
Other languages
English (en)
French (fr)
Inventor
Nicholas M/S R338 VALIANTE
Feng M/S R338 XU
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.)
Novartis Vaccines and Diagnostics Inc
Original Assignee
Novartis Vaccines and Diagnostics Inc
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 Novartis Vaccines and Diagnostics Inc filed Critical Novartis Vaccines and Diagnostics Inc
Priority to EP10179748A priority Critical patent/EP2277595A3/de
Publication of EP1765313A2 publication Critical patent/EP1765313A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • A61K31/708Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid having oxo groups directly attached to the purine ring system, e.g. guanosine, guanylic acid
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention pertains generally to compounds capable of stimulating or modulating an immune response in a subject.
  • the compounds are 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-diones, staurosporine analogs, derivatized pyridazines, cliromen-4-ones, indolinones, quinazolines, nucleoside analogs, or other small molecules as described herein.
  • the invention provides novel combinations of antigens with immune potentiators that may be used in vaccine therapies.
  • the compounds in one embodiment can be used as immunotherapeutics for proliferative diseases, infectious diseases, autoimmune diseases and/or allergies/asthma.
  • the maximum tolerated doses (MTDs) necessary for treatment efficacy may result in undesirable side effects and even weaken the immune response in the patient. Such side effects may require cessation of treatment.
  • MTDs maximum tolerated doses
  • the dual action of inhibiting bcr-abl, while stimulating an immune response likely contributes to its efficacy and tolerability, particularly because NK cells, which are stimulated by administration of Gleevec®, independently play a role in tumor recession. This synergistic approach to cancer regression is extremely effective.
  • cytotoxics that suppress the immune system may independently contribute to the disease state since they may inhibit separate pathways that are involved in recovery.
  • Another advantage to immune potentiation is that it provides a platfo ⁇ n less easily bypassed by resistance mutations.
  • therapeutic targets are so polarized and specific (which may be necessary in order to avoid targeting host cells), such as a particular substrate in a viral replicon or a kinase in a cancer cell line
  • a single point mutation in the disease state may render it unaffected by a drug resulting in even harsher strains of the disease in future generations.
  • Novel methods and mechanisms for treating patients having diseases resistant to, or inadequately treated by, conventional approaches utilizing agents targeting specific immune response mechanisms in the body is needed.
  • Substances that stimulate immune cells in vitro exhibit similar immuno- stimulatory effects in vivo.
  • cytokines such as recombinant cytokines, pathogen products (e.g. toxins, lipids, proteins/peptides, carbohydrates and nucleic acids) and other mammalian-derived immunostimulatory molecules (e.g. heat shock proteins, immune complexes and proteoglycans) all induce a measurable pro ⁇ inflammatory response both in vitro and in vivo.
  • pathogen products e.g. toxins, lipids, proteins/peptides, carbohydrates and nucleic acids
  • immunostimulatory molecules e.g. heat shock proteins, immune complexes and proteoglycans
  • the classic adjuvants have been Freund's complete or incomplete (i.e., without mycobacteria) adjuvants.
  • Edmund Coley described the potential of Coley's toxin for cancer immuno-therapy.
  • Other materials such as mineral oil and aluminum hydroxide, have also been used as adjuvants, but they invariably suffer from disadvantages.
  • mineral oil is known to produce tissue irritation and to be potentially oncogenic.
  • Alum the only approved adjuvant in the United States, also induces granulomas at the inoculation site and furthermore it does not effectively induce cell-mediated immunity.
  • many of the adjuvants currently available have limited utility because they contain components that are not metabolizable in humans.
  • cytotoxic drugs are typically only effective against cancers with rapid growth rates, limiting their applicability and effective patient populations.
  • Compounding the problem of toxicity and weakened immune response is the potential for an increased fraction of treatment resistant carcinogenic cells as therapy progresses. In order for treatment to be successful (beyond palliative therapy), it is necessary that all of the cancer cells be eliminated from the patient. Where the drug is so selective in its targeted mechanism of action, the likelihood of drug resistance increases since cancer cell survival depends solely on modifying a single targeted point in the cellular machinery.
  • Drug regimens have been designed to kill as many tumor cells as possible by treating with "maximum tolerated doses" (MTDs) of these cytotoxic agents.
  • MTDs maximum tolerated doses
  • a conventional dosing schedule calls for episodic application of a cytotoxic drug at or near the MTD, followed by periods of rest to allow normal tissues to recover.
  • Many such chemotherapy regimens are initially efficacious, resulting in tumor regression, but eventually lead relapses often marked by aggressive cancers that are resistant to the cytotoxic drug.
  • an altered regimen with multiple mechanisms of action, functioning at doses low enough to avoid toxicity, while stimulating an immune response would be beneficial.
  • Such a regimen could be used in patients with both rapidly growing tumor cell lines as well as those with low growth rates and/or those in remission to prevent a relapse, since toxicities would be low and immune system functionality would be enhanced as opposed to being suppressed.
  • the current invention also seeks to provide individual therapeutic and prophylactic agents for treatment of disease states characterized by other immune deficiencies or abnormalities, including autoimmune diseases responsive to compounds with the capacity to modulate cytokines and/or TNF-a, such as multiple sclerosis and Crohn's disease.
  • SMIPs small molecule immune potentiators
  • the small molecule platform provides diverse compounds for the selective manipulation of the immune response, necessary for increasing the therapeutic index immune modulators.
  • novel compounds with a varied capacity to alter levels and/or profiles of cytokine production in human immune cells Compounds with structural disparities will often times elicit a desired response through a different mechanism of action, or with greater specificity to a target, such as a dendritic cell, modulating potency and lowering side effects when administered to a patient.
  • a target such as a dendritic cell, modulating potency and lowering side effects when administered to a patient.
  • novel sole acting agents with a varied capacity to alter levels and/or profiles of cytokine production in human immune cells.
  • chemotherapies may be offered that combine increasing efficiency with a large reduction of side effects and therapeutic doses.
  • the therapeutic efficiency of known cytostatic drugs is increased.
  • certain cell lines that are insensitive to chemotherapeutic treatment may become susceptible to chemotherapy by applying the combination of active substances.
  • the current invention also seeks to provide individual therapeutic and prophylactic agents for treatment of disease states characterized by other immune deficiencies, abnormalities, or infections including autoimmune diseases and viral and bacterial infections responsive to compounds with the capacity to modulate cytokines and/or TNF-a, such as multiple sclerosis, Crohn's disease, HIV, HSV, and HCV, among others.
  • Therapeutics that could serve to augment natural host defenses against viral infections with reduced toxicity would be very beneficial.
  • the present invention provides such therapeutic agents, and further provides other related advantages.
  • the present invention provides immunogenic compositions and novel methods of administering a vaccine by administering 3,4-di(lH-indol-3-yl)-lH- pyrrole-2,5-diones in combination with antigens or other agents.
  • the invention further provides 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5- diones as pharmaceutical compositions, for use in the treatment of cancer, infectious diseases, allergies, and asthma. More particularly the invention provides 3,4-di(lH- indol-3-yl)-lH-pyrrole-2,5-diones for treatment of diseases in patients with suppressed immune systems.
  • 3,4-di(lH- indol-3-yl)-lH-pyrrole-2,5-diones surpass existing therapies in that they function by independently targeting substrates involved in cancer pathways, while separately inducing an immune response in the subject thereby attacking different modes of the disease state.
  • the 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds are combined with numerous antigens and delivery systems to form a final vaccine product.
  • the 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds are used alone or in combination with other therapies (e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of chronic infections such as HIV, HCV, HBV, HSV, and H. pylori, as well as medicaments for the reduction of tumor growth.
  • other therapies e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens
  • the 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds also may be used for the treatment of cancer either alone or in combination with other anti-cancer therapies (e.g.
  • chemotherapeutic agents mAbs or other immune potentiators.
  • certain 3,4-di(lH-indol-3-yl)-lH-pyrrole- 2,5-diones with the capacity to induce Type 1 cytokines (e.g. IL- 12, TNF or IFN 's) could be useful for the treatment of allergies or asthma due to their capacity to steer the immune response towards more benign sequelae.
  • Type 1 cytokines e.g. IL- 12, TNF or IFN 's
  • the 3,4-di(lH-indol-3-yl)-lH- pyrrole-2,5-dione compounds may be used for example for the treatment of BCG, cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenzae b, meningococcus infection, and pneumococcus infection.
  • the 3,4-di(lH- indol-3-yl)-lH-pyrrole-2,5-dione compounds may be used in an anti cell proliferative effective amount for the treatment of cancer.
  • the 3,4-di(lH-indol-3-yl)-lH-pyrrole- 2,5-dione compounds may also be used in anti-Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • methods of treating cancer are provided wherein known anticancer agents are combined with 3,4- di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds to reduce tumor growth in a subject.
  • a method of inhibiting tumor cell growth comprising administering to a subject an effective dose of a combination containing at least one 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione and a mAb or antigen, wherein said combination is more effective to inhibit such cell growth than when said mAb or antigen is administered individually.
  • the present invention provides immunogenic compositions and novel methods of administering a vaccine by administering indolinones in combination with antigens or other agents.
  • the invention further provides indolinones as pharmaceutical compositions, for use in the treatment of cancer, infectious diseases, allergies, and asthma. More particularly the invention provides indolinones for treatment of diseases in patients with suppressed immune systems.
  • indolinones surpass existing therapies in that they function by independently targeting substrates involved in cancer pathways, while separately inducing an immune response in the subject thereby attacking different modes of the disease state.
  • the indolinone compounds are combined with numerous antigens and delivery systems to form a final vaccine product.
  • the indolinone compounds are used alone or in combination with other therapies (e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of chronic infections such as HIV, HCV, HBV, HSV, and H. pylori, as well as medicaments for the reduction of tumor growth.
  • the indolinone compounds also may be used for the treatment of cancer either alone or in combination with other anti-cancer therapies (e.g. chemotherapeutic agents, mAbs or other immune potentiators).
  • indolinones with the capacity to induce Type 1 cytokines (e.g. IL-12, TNF or IFN's) could be useful for the treatment of allergies or asthma due to their capacity to steer the immune response towards more benign sequelae.
  • the indolinone compounds may be used for example for the treatment of BCG, cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenzae b, meningococcus infection, and pneumococcus infection.
  • the indolinone compounds may be used in an anti cell proliferative effective amount for the treatment of cancer.
  • the indolinone compounds may also be used in anti-Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • methods of treating cancer comprising administering to a subject an effective dose of a combination containing at least one indolinone and a mAb or antigen, wherein said combination is more effective to inhibit such cell growth than when said mAb or antigen is administered individually.
  • Preferred SMIPs of the second aspect of the present invention include indolinone compositions, specifically N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2- oxoindolin-3-ylidene)methyl)-2,4-dimethyl-lH-pyrrole-3-carboxamide; 3-((3,5- dimethyl-lH-pyrrol-2-yl)methylene)indolin-2-one; 3-(2,4-dimethyl-5-((2-oxoindolin- 3-ylidene)methyl)-lH-pyrrol-3-yl)propanoic acid; and 3-(3,5-dibromo-4- hydroxybenzylidene) -5 -iodoindolin-2-one .
  • the present invention provides immunogenic compositions and novel methods of administering a vaccine by administering chromen-4-ones in combination with antigens or other agents.
  • the third aspect of the invention further provides chromen-4-ones as pharmaceutical compositions, for use in the treatment of cancer, infectious diseases, allergies, and asthma. More particularly the invention provides chromen-4-ones for treatment of diseases in patients with suppressed immune systems.
  • chromen-4-ones surpass existing therapies in that they function by independently targeting substrates involved in cancer pathways, while separately inducing an immune response in the subject thereby attacking different modes of the disease state.
  • the chromen-4-one compounds are combined with numerous antigens and delivery systems to form a final vaccine product.
  • the chromen-4-one compounds are used alone or in combination with other therapies (e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of chronic infections such as HIV, HCV, HBV, HSV, and H. pylori, as well as medicaments for the reduction of tumor growth.
  • chromen-4-one compounds also may be used for the treatment of cancer either alone or in combination with other anti-cancer therapies (e.g. chemotherapeutic agents, mAbs or other immune potentiators).
  • chemotherapeutic agents e.g. chemotherapeutic agents, mAbs or other immune potentiators.
  • certain chromen-4-ones with the capacity to induce Type 1 cytokines e.g. IL-12, TNF or IFN' s
  • Type 1 cytokines e.g. IL-12, TNF or IFN' s
  • the chromen-4-one compounds may be used for example for the treatment of BCG, cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenzae b, meningococcus infection, and pneumococcus infection.
  • the chromen-4-one compounds may be used in an anti cell proliferative effective amount for the treatment of cancer.
  • the chromen-4-one compounds may also be used in anti- Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • methods of treating cancer wherein known anticancer agents are combined with chromen-4-one compounds to reduce tumor growth in a subject.
  • a method of inhibiting tumor cell growth comprising administering to a subject an effective dose of a combination containing at least one chromen-4-one and a mAb or antigen, wherein said combination is more effective to inhibit such cell growth than when said mAb or antigen is administered individually.
  • Preferred SMIPs of the third aspect of the present invention include chromen- 4-one compositions, specifically 2-(2-chlorophenyl)-2,3-dihydro-5,7-dihydroxy-8-(3- hydroxy-l-methylpiperidin-4-yl)chromen-4-one; 5,7-dihydroxy-3-(4-hydroxyphenyl)- 4H-chromen-4-one; and 2,3 -dihydro-5,7-dihydroxy-6-methoxy-2-(3 ,A- dimethoxyphenyl)chromen-4-one.
  • the present invention provides immunogenic compositions and novel methods of administering a vaccine by administering derivatized pyridazines in combination with antigens or other agents.
  • the invention further provides derivatized pyridazines as pharmaceutical compositions, for use in the treatment of cancer, infectious diseases, allergies, and asthma.
  • the invention provides derivatized pyridazines for treatment of diseases in patients with suppressed immune systems.
  • derivatized pyridazines surpass existing therapies in that they function by independently targeting substrates involved in cancer pathways, while separately inducing an immune response in the subject thereby attacking different modes of the disease state.
  • the derivatized pyridazine compounds are combined with numerous antigens and delivery systems to form a final vaccine product.
  • the derivatized pyridazine compounds are used alone or in combination with other therapies (e.g.
  • the derivatized pyridazine compounds also may be used for the treatment of cancer either alone or in combination with other anti-cancer therapies (e.g. chemotherapeutic agents, niAbs or other immune potentiators).
  • certain derivatized pyridazines with the capacity to induce Type 1 cytokines e.g.
  • the derivatized pyridazine compounds may be used for example for the treatment of BCG, cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenzae b, meningococcus infection, and pneumococcus infection.
  • the derivatized pyridazine compounds may be used in an anti cell proliferative effective amount for the treatment of cancer.
  • the derivatized pyridazine compounds may also be used in anti-Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • methods of treating cancer comprising administering to a subject an effective dose of a combination containing at least one derivatized pyridazine and a mAb or antigen, wherein said combination is more effective to inhibit such cell growth than when said mAb or antigen is administered individually.
  • Preferred SMIPs of the fourth aspect of the present invention include derivatized pyridazine compositions, specifically l-(4-Chloroanilino)-4-(4- pyridylmethyl)phthalazine.
  • Additional embodiments, methods and compositions contemplated to be useful in the fourth aspect of the instant invention are disclosed in USSN 10/814,480, 10/762,873, 60/582,654, US 6,258,812 and 10/748,071 which are incorporated by reference as if set forth fully herein.
  • the present invention provides immunogenic compositions and novel methods of administering a vaccine by administering staurosporine analogs in combination with antigens or other agents.
  • the fifth aspect of the invention further provides staurosporine analogs as pharmaceutical compositions, for use in the treatment of cancer, infectious diseases, allergies, and asthma. More particularly the invention provides staurosporine analogs for treatment of diseases in patients with suppressed immune systems. For the treatment of cancer at lowered doses, staurosporine analogs surpass existing therapies in that they function by independently targeting substrates involved in cancer pathways, while separately inducing an immune response in the subject thereby attacking different modes of the disease state. [0052] As adjuvants, the staurosporine analog compounds are combined with numerous antigens and delivery systems to form a final vaccine product. [0053] As immuno-therapeutics, the staurosporine analog compounds are used alone or in combination with other therapies (e.g.
  • the staurosporine analogs also may be used for the treatment of cancer either alone or in combination with other anti-cancer therapies (e.g. chemotherapeutic agents, niAbs or other immune potentiators).
  • certain staurosporine analogs with the capacity to induce Type 1 cytokines e.g. IL- 12, TNF or IFN 's
  • Type 1 cytokines e.g. IL- 12, TNF or IFN 's
  • the staurosporine analog compounds may be used for example for the treatment of BCG, cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenzae b, meningococcus infection, and pneumococcus infection.
  • the staurosporine analog compounds may be used in an anti cell proliferative effective amount for the treatment of cancer.
  • the staurosporine analog compounds may also be used in anti-Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • methods of treating cancer wherein known anticancer agents are combined with staurosporine analog compounds to reduce tumor growth in a subject.
  • a method of inhibiting tumor cell growth comprising administering to a subject an effective dose of a combination containing at least one staurosporine analog and a mAb or antigen, wherein said combination is more effective to inhibit such cell growth than when said mAb or antigen is administered individually.
  • Preferred SMIPs of the fifth aspect of the present invention include staurosporine analog compositions, specifically 9,13-Epoxy-lH,9H-diindolo[l,2,3- gh ⁇ ' ⁇ U'-Mpyrrolop ⁇ fl ⁇ benzodiazonin-l-one, 2,3,10,11,12,13-hexahydro- 11,12-dihydroxy- 10-methoxy-9-methyl; 9,13 -Epoxy- 1 H,9H-diindolo[ 1 ,2,3-gh: 3 ⁇ 2 ⁇ Y- lm]pyrrolo[3 ,4-j] [ 1 ,7]benzodiazonin-l -one, 2,3 , 10, 11 , 12, 13-hexahydro- 10-methoxy- 9-methyl-ll-(methylamino); 9,13-Epoxy-lH,9H-diindolo[l,2,3-gh:3',2',r-
  • the instant invention provides novel immune potentiators, immunogenic compositions, and novel methods of administering a vaccine, by administering nucleoside analogs alone or in combination with antigens/other agents.
  • the sixth aspect of the invention further provides compositions, novel compounds and pharmaceutical compositions, for use in the treatment of infectious diseases.
  • the nucleoside analogs used in the methods and compositions of the sixth aspect of the invention are inexpensive to produce and easy to administer.
  • the nucleoside analogs are combined with numerous antigens and delivery systems to form a final vaccine product.
  • the nucleoside analogs are used alone or in combination with other therapies (e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of chronic infections such as Kaposi's sarcoma, anogenital warts, HIV, HCV, SARS, HBV, HPV, HSV, and H. pylori.
  • nucleoside analogs of the present invention target substrates in the virus or disease state, such as, for example proteases, replicases, DNA polymerase, and/or RNA polymerase.
  • substrates in the virus or disease state such as, for example proteases, replicases, DNA polymerase, and/or RNA polymerase.
  • nucleoside analogs with the capacity to induce Type 1 cytokines e.g. IL-12, TNF or IFN's
  • Type 1 cytokines e.g. IL-12, TNF or IFN's
  • the nucleoside analogs may be used for example for the treatment of BCG, cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenzae b, meningococcus infection, and pneumococcus infection.
  • the nucleoside analogs may be used in an anti cell proliferative effective amount for the treatment of cancer.
  • the nucleoside analogs may also be used in anti-Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • the present invention immunogenic compositions and novel methods of administering a vaccine, by administering fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD- 6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-Ol, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, or tetrathiomolybdate in combination with antigens or other agents.
  • the seventh aspect of the invention further provides one of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP- 701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY- 317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, or tetrathiomolybdate as a pharmaceutical composition, for use in the treatment of diseases associated or complicated with immune suppression, such as cancer, infectious diseases, allergies, and asthma.
  • diseases associated or complicated with immune suppression such as cancer, infectious diseases, allergies, and asthma.
  • the SMIP compounds namely fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN- 518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, or tetrathiomolybdate) are combined with numerous antigens and delivery systems to fo ⁇ n a final vaccine product.
  • the SMIP compounds namely fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R
  • the SMIP compounds are used alone or in combination with other therapies (e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of autoimmune diseases, chronic infections such as HIV, HCV, HBV, HSV, and H. pylori, as well as medicaments for the reduction of tumor growth.
  • therapies e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens
  • autoimmune diseases e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens
  • chronic infections such as HIV, HCV, HBV, HSV, and H. pylori
  • medicaments for the reduction of tumor growth e.g. anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens
  • one embodiment of the seventh aspect of the invention provides administering a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP- 701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY- 317615, squalamine, UCN-Ol, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate to treat one of the following diseases: bacterial diseases autoimmune-, allergic- and viral-diseases, disturbances caused by the incidence of mental diseases, central nervous system depressants, habitual alcohols, and disorders of the respiratory system
  • the SMIP compounds also may be used for the treatment of cancer either alone or in combination with other anti-cancer therapies (e.g. chemotherapeutic agents, mAbs or other immune potentiators).
  • chemotherapeutic agents e.g. chemotherapeutic agents, mAbs or other immune potentiators.
  • IL-12, TNF or IFN's are useful for the treatment of allergies or asthma due to their capacity to steer the immune response towards more benign sequelae.
  • methods of treating cancer comprising administering to a subject an effective dose of a combination containing at least one SMIP and a Mab or antigen, wherein said combination is more effective to inhibit such cell growth than when said Mab or antigen is administered individually.
  • Preferable SMIPs of the seventh aspect of the present invention include fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, or tetrathiomolybdate compositions as well as analogs disclosed in the following patents and patent applications: US 4,323,581, US 6,258,812, WO 98/35958, WO 01/60814, US 5,883,113, WO 99/61422, US 5,883,113, WO 99
  • preferred compounds of the seventh aspect include those encompassed by Formula I in the aforementioned references.
  • Additional embodiments, methods and compositions contemplated to be useful in the seventh aspect of the instant invention are disclosed in USSN 10/814,480, 10/762,873, and 10/748,071.
  • Further embodiments of the several aspects of the invention include those described in the detailed description.
  • the "method of modulating an immune response in a patient comprising administering a compound" of a particular formula, such as formula (I) can be replaced with: "use of a compound of formula (I) in the manufacture of a medicament for modulating an immune response in a patient.”
  • the compounds are used in the manufacture of a medicament for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the compounds are used in the manufacture of a medicament for use as an adjuvant.
  • Preferred compounds of the present invention include those having a particular formula as described in the aspects of the invention.
  • Other embodiments provide the use of a compound of formula (I) and another agent for simultaneous separate or sequential administration.
  • the other agent is an antigen.
  • the use is for modulating an immune response in a patient.
  • the use is for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the use is as an adjuvant.
  • Other embodiments provide a pharmaceutical preparation or system, comprising (a) a first pharmaceutical agent, which comprises a compound of formula (I); and (b) a second pharmaceutical agent, wherein said first and second agents are either in admixture or are separate compositions.
  • the second agent is an antigen. More specifically, the agents are for simultaneous separate or sequential administration.
  • the use is for modulating an immune response in a patient.
  • kits comprising (a) a first pharmaceutical agent, which comprises a SMIP of formula I-L; and (b) a second pharmaceutical agent.
  • the second agent is an antigen.
  • the use is for modulating an immune response in a patient.
  • the use is for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the use is as an adjuvant.
  • Another embodiment provides the use of a compound of formula (I) and another agent in the manufacture of a combination medicament.
  • the other agent is an antigen.
  • the use is for modulating an immune response in a patient.
  • the use is for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the use is as an adjuvant.
  • Another embodiment provides the use of a compound of formula (I) in the manufacture of a medicament, wherein the medicament is co-administered with another agent.
  • the second agent is an antigen.
  • the use is for modulating an immune response in a patient.
  • the use is for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the use is as an adjuvant.
  • Another embodiment provides the use of an antigen in the manufacture of a medicament, wherein the medicament is co-administered with a compound of formula (I).
  • the two agents are preferably administered within 4 hours of each other.
  • Another embodiment provides the use of a compound of formula (I) in the manufacture of a medicament, wherein the medicament is for administration to a patient who has been pre-treated with another agent.
  • the second agent is an antigen.
  • the use is for modulating an immune response in a patient.
  • the use is for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the use is as an adjuvant.
  • Another embodiment provides the use of an antigen in the manufacture of a medicament, wherein the medicament is for administration to a patient who has been pre-treated with a compound of formula (I).
  • the pre-treatment may be recent (e.g. within the 24 hours preceding administration of said medicament), intermediate (e.g. more than 24 hours previous, but no longer than 4 weeks), more distant (e.g. at least 4 weeks previous), or very distant (e.g. at least 6 months previous), with these time periods referring to the most recent pre treatment dose.
  • the patient may be refractory to treatment by the pharmaceutical agent that was administered in the pre- treatment.
  • the use is for modulating an immune response in a patient.
  • the use is for treating an infectious disease, autoimmune disease, allergies, or cancer.
  • the use is as an adjuvant.
  • Another embodiment provides, the use of a compound of formula (I) in the manufacture of a medicament, wherein the medicament is for administration to a patient who has a tumor or infection that is resistant to treatment with another agent.
  • One embodiment of the first aspect invention includes a method of modulating an immune response in a subject comprising administering a compound of fo ⁇ nula I: I wherein, Ri and R 2 are each independently H, alkyl, aryl, alkoxy, -C(O)R 0 , or heterocyclyl; or Ri and R 2 are taken together to form a bridge in subformula Ia:
  • R 3 and R 4 each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R c , -S(O) p R d , or heterocyclyl;
  • R 5 is H or alkyl;
  • R 6 is H, -OH, -NH 2 , halogen, or alkyl;
  • R 7 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R 0 , -S(O) p R d , or heterocyclyl;
  • R 5 is H.
  • R 3 and R 4 are both H.
  • Ri is methyl, ethyl, propyl, or isopropyl
  • R 2 is heterocyclyl.
  • R 2 is defined as in subformula R 2a : the binding being achieved at the bond crossed with a -TM ⁇ ; Z is N, O, S, or CH, provided that when Z is O or S, w is 0 and R 8 is absent; R 8 is absent, aryl or heterocyclyl; v is 0, 1, or 2; and w is 0, 1, or 2.
  • Z is N.
  • w and v are both 1.
  • R 8 is pyridyl.
  • Rj and R 2 are taken together to form a bridge in subformula Ia:
  • R 6 is H.
  • n is 1.
  • X is O.
  • R 7 is -CH 2 -NH(alkyl), -CH 2 - NH 2 , -CH 2 -CH 2 -NH 2 , -CH 2 -CH 2 -NH(alkyl), -CH 2 -CH 2 -N(alkyl) 2 or -CH 2 - N(alkyl) 2 .
  • alkyl with R 7 is methyl, ethyl, propyl, or isopropyl.
  • X is -CH 2 -.
  • R 7 is -CH 2 -NH(alkyl), -CH 2 - NH 2 , -CH 2 -CH 2 -NH 2 , -CH 2 -CH 2 -NH(alkyl), -CH 2 -CH 2 -N(alkyl) 2 or -CH 2 -N(alkyl) 2 .
  • alkyl with R 7 is methyl, ethyl, propyl, or isopropyl.
  • said modulating is inducing.
  • said inducing stimulates production of cytokines, chemokines, and/or growth factors.
  • said compound is administered in a subcytotoxic amount to said subject.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • the subject is not suffering from cancer.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF-ce levels.
  • said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • subject is suffering from an autoimmune disease. Further still, said autoimmune disease is multiple sclerosis or Crohn's disease.
  • said subject is suffering from a viral infection.
  • said viral infection is HCV, HIV, or HSV.
  • said subject is suffering from allergies.
  • said subject is suffering from asthma.
  • precancerous lesions such as actinic keratosis, atypical or dysplastic nevi, or premalignant lentigos.
  • the subject is suffering from a disease associated with abnormal cellular proliferation, such as, neuro-f ⁇ bromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • a disease associated with abnormal cellular proliferation such as, neuro-f ⁇ bromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • Another embodiment provides a pharmaceutical composition containing any of the aforementioned compounds or embodiments of formula I.
  • the subject is suffering from precancerous lesions
  • the 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds can be used with or without an antigen in therapeutic applications, for example to treat cancer or infectious diseases.
  • the 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds also may be used in combination with other therapeutic agents, such as anti-virals and monoclonal antibodies in different therapeutic applications.
  • One preferred embodiment of the method of inducing an immunostimulatory effect in a patient is directed to administering an immunogenic composition comprising a vaccine in an amount effective to stimulate an immune response such as a cell-mediated immune response and, as a vaccine adjuvant, an 3,4- di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compound, in an amount effective to potentiate the immune response such as the cell-mediated immune response to the vaccine.
  • Agents combined with the 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compounds, contemplated to be useful in treating the aforementioned diseases include those well known in the art, such as, anesthetics, hypnotic sedatives, anti-anxieties, antiepileptics, antipyretic antiphlogistics, stimulants, wake amines, anti-parkinson drugs, agents for psychoneuroses, agents for central nervous system, skeletal muscle relaxants, agents for autonomic nervous system, antispastic agents, cytotoxic agents, monoclonal antibodies, drugs for eye, drugs for nose and ear, anti-vertiginous drugs, cardiotonics, antiarrhythmic drugs, diuretics, pressure reduction drugs, vasoconstrictors, coronary vaso-dilators, peripheral vasodilating drugs, hyper-lipemia drugs, breath stimulants, antitussive and expectorant drugs, bronchodil
  • compositions described herein are used for the treatment of cancer and reduction of tumor growth.
  • an 3,4-di(lH- indol-3-yl)-lH-pyrrole-2,5-dione compound of the invention is combined with a known mAb for the treatment of cancer.
  • an antibody and a 3,4-di(lH-indol-3-yl)-lH- pyrrole-2,5-dione compound are administered.
  • a therapeutic composition for inhibiting tumor cell growth in a subject which composition comprises an effective amount of a combination of at least an 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-dione compound and a mAb and a pharmaceutically acceptable carrier, wherein said combination is more effective to inhibit growth of certain mammalian tumor cells than are either of the agents when administered individually.
  • methods of treating allergies comprising administering an 3,4-di(lH-indol-3-yl)-lH- pyrrole-2,5-dione compound alone or in combination with another agent known to be effective against allergies, wherein said combination is more effective in treating an allergic condition than the known agent(s) are without the addition of said 3,4-di(lH- indol-3-yl)-lH-pyrrole-2,5-dione compound.
  • the known agent is an antihistamine and/or a leukotriene inhibitor.
  • the allergic condition is asthma.
  • the allergic condition is selected from the group consisting of allergic rhinitis, dermatosis, and urticaria.
  • the combination is administered to a subject enterally, parenterally, intranasally, subcutaneously, or intraarterially.
  • Preferred SMIPs in accordance with the first aspect of the invention are 3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-diones, as well as analogs disclosed in the following patents and patent applications: US 5,552,396, US 5,057,614, and WO 95/17182.
  • preferred SMIPs of the first aspect of the invention include flavones, isoflavones and those encompassed by Formula I as described in this section, or contained within the aforementioned references.
  • Reference to "3,4-di(lH-indol-3-yl)-lH-pyrrole-2,5-diones" indicates compounds having the general structure of Formula I as described herein.
  • Preferred 3 ,4-di( 1 H-indol-3 -yl)-l H-pyrrole-2,5-diones are [00125] The foregoing may be better understood by reference to the Examples, infra, that are presented for illustration and not to limit the scope of the inventive concepts.
  • Example compounds and their analogs are either commercially available, or are easily synthesized by one skilled in the art from procedures described in patents/applications, such as US 5,552,396, US 5,057,614, WO 95/17182 and other patents/applications listed herein.
  • Section K Second Aspect of the Invention - Indolinones for Immunopotentiation
  • AU definitions, descriptors of constituent variables for chemical formulas, and descriptions appearing in the section shall be understood to apply to this section only.
  • Applicants have discovered methods of stimulating cytokine activity in cells and immunotherapeutics and/or vaccine adjuvants, that will provide effective treatments for disorders described herein and those apparent to one skilled in the art.
  • a method of modulating an immune response in a subject comprising administering a compound of formula I: I wherein, Ri is alkyl, aryl or heterocyclyl; R 2 , R 3 , R 4 , and R 5 are each independently H, -CN, -OH 3 halogen, alkyl, aryl, alkoxy, -NR 3 Rb, -C(O)Rc, -S(O) n Rd, or heterocyclyl; each R a and R b is independently H, alkyl, -C(O)R 0 , aryl, heterocyclyl, or alkoxy; each R c is independently H, alkyl, alkoxy, -NH 2 , -NH(alkyl), -N(alkyl) 2 , aryl, or heterocyclyl; each R d is independently H, alkyl, alkenyl, aryl, or -NR
  • Ri is heterocyclyl. In a more particular embodiment still, Ri is R] a :
  • R 6 , R 7 and Rg are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, - NR a Rb, -C(O)R 0 , -S(O) n Rd, or heterocyclyl.
  • Ri is R] a
  • R 7 is -C(O)-(CH 2 ) p -N(H)( 2-r) (alkyl) r , wherein p is 0, 1, 2, 3, 4, or 5 and r is 0, 1, or 2.
  • Ri Ria
  • R 3 is F.
  • Ri is Ri a
  • R 7 is - (CH 2 ) t COOH, wherein t is 1, 2, 3, or 4.
  • Ri is Ri 2
  • R 6 and R 8 are both methyl.
  • Rj is R] a
  • R 7 is H.
  • Ri is aryl. More particular still, Ri is substituted or unsubstituted phenyl.
  • R 3 is iodide.
  • said modulating is inducing.
  • said inducing stimulates production of cytokines, chemokines, and/or growth factors.
  • said compound is administered in a subcytotoxic amount to said subject.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • the subject is not suffering from cancer.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels.
  • said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • subject is suffering from an autoimmune disease. Further still, said autoimmune disease is multiple sclerosis.
  • said subject is suffering from a viral infection.
  • said viral infection is HCV, HIV, or HSV.
  • subject is suffering from allergies.
  • said subject is suffering from asthma. [00146] It is contemplated that the invention encompasses all possible combinations of the embodiments described herein.
  • Preferred SMIPs in accordance with the second aspect of the invention are indolinones, such as N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3- ylidene)methyl)-2,4-dimethyl-lH-pyrrole-3-carboxamide; 3-((3,5-dimethyl-lH- pyrrol-2-yl)methylene)mdolin-2-one; 3-(2,4-dimethyl-5-((2-oxoindolin-3- ylidene)methyl)-lH-pyrrol-3-yl)propanoic acid; and 3-(3,5-dibromo-4- hydroxybenzylidene)-5-iodoindolin-2-one, as well as analogs disclosed in the following patents and patent applications: WO 01/60814, US 5,883,113, WO 99/61422, and WO 99/61422.
  • preferred SMIPs include those encompassed by Formula I as described in this section, or contained within the aforementioned references.
  • Reference to "indolinones” indicates compounds having the general structure of Fo ⁇ nula I as described in this section.
  • Preferred indolinones are N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-dimethyl-lH- pyrrole-3-carboxamide; 3-((3,5-dimethyl-lH-pyrrol-2-yl)methylene)indolin-2-one; 3- (2,4-dimethyl-5-((2-oxoindolin-3-ylidene)methyl)-lH-pyrrol-3-yl)propanoic acid; and 3 -(3 , 5-dibromo-4-hydiOxybenzylidene)-5 -iodoindolin-2-one .
  • Section III Third Aspect of the Invention - Chromen-4-ones for Immiinopotentiation
  • AU definitions, descriptors of constituent variables for chemical formulas, and descriptions appearing in the section shall be understood to apply to this section only.
  • Applicants have discovered methods of stimulating cytokine activity in cells and immunotherapeutics and/or vaccine adjuvants, that will provide effective treatments for disorders described herein and those apparent to one skilled in the art.
  • a method of modulating an immune response in a subject comprising administering a compound of formula I:
  • Ri and R 2 is H, -CN, alkyl, -OH, -NR a R b , alkoxy, -C(O)Rc, -S(O) n Rd, heterocyclyl, or aryl, and the other is:
  • R 3 , R 4 , R 5 , and R 6 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)Rc, -S(O) n Rd, or heterocyclyl;
  • R 7 and Ri 0 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R 0 , -S(O) n Rd, or heterocyclyl;
  • R 8 and R 9 are each independently H, -CN, -OH, halogen, alkyl, aryl, -O-(CH 2 ) q -R e , -C(O)Rc, -0-(CH 2 ) q -0-R e , -NR a R b , -S(O) n R d , or hetero
  • R 3 is heterocyclyl. [00157] In another more particular embodiment thereof, R 3 is:
  • R 7 is Cl.
  • R 4 and R 6 are both -OH.
  • R 5 is alkoxy.
  • R 5 is methoxy.
  • R 8 and R 9 are both -O-(CH 2 ) q - Rc
  • R 8 and Rg are both -OCH 3 .
  • R 2 is H. [00165] In another embodiment R 2 is:
  • Ri is H.
  • R 4 and R 6 are both -OH.
  • R 9 is -OH.
  • Another embodiment of the invention provides a method of modulating an immune response in a subject comprising administering a compound selected from the group consisting of:
  • said modulating is inducing.
  • said inducing stimulates production of cytokines, chemokines, and/or growth factors.
  • said compound is administered in a subcytotoxic amount to said subject.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels.
  • said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • subject is suffering from an autoimmune disease. Further still, said autoimmune disease is multiple sclerosis or Crohn's disease.
  • said subject is suffering from a viral infection.
  • said viral infection is HCV, HIV, or HSV.
  • said subject is suffering from allergies.
  • said subject is suffering from asthma.
  • the subject is suffering from a disease associated with abnormal cellular proliferation, such as, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • a disease associated with abnormal cellular proliferation such as, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • Preferred SMIPs in accordance with the third aspect of the invention are chromen-4-ones, such as 2-(2-chlorophenyl)-2,3-dihydro-5,7-dihydroxy-8-(3- hydroxy- 1 -methylpiperidin-4-yl)chrornen-4-one; 5 ,7-dihydroxy-3 -(4-hydroxyphenyl)- 4H-chromen-4-one; and 2,3-dihydro-5,7-dihydroxy-6-methoxy-2-(3,4- dimethoxyphenyl)chromen-4-one, as well as analogs disclosed in the following patents and patent applications: WO 97/42949, and WO 98/13344, US 5,554,519, WO 98/04541, and US 6,025,387.
  • chromen-4-ones such as 2-(2-chlorophenyl)-2,3-dihydro-5,7-dihydroxy-8-(3- hydroxy- 1 -methylpiperidin-4-y
  • preferred SMIPs include flavones, isoflavones and those encompassed by Formula I as described in this section, or contained within the aforementioned references.
  • Reference to "chromen-4-ones” indicates compounds having the general structure of Formula I as described in this section.
  • Preferred chromen-4-ones are flavones and isoflavones selected from the group consisting of 2-(2- chlorophenyl)-2,3-dihydro-5,7-dihydroxy-8-(3-hydroxy-l-methylpiperidin-4- yl)chromen-4-one; 5,7-dihydroxy-3-(4-hydroxyphenyl)-4H-chromen-4-one; and 2,3- dihydro-5,7-dihydroxy-6-methoxy-2-(3,4-dimethoxyphenyl)chromen-4-one.
  • Section IV Fourth Aspect of the Invention - Derivatized Pyridazines for Immunopotentiation
  • All definitions, descriptors of constituent variables for chemical formulas, and descriptions appearing in the section shall be understood to apply to this section only.
  • Applicants have discovered methods of stimulating cytokine activity in cells and immunotherapeutics and/or vaccine adjuvants, that will provide effective treatments for disorders described herein and those apparent to one skilled in the art.
  • One embodiment of the invention provides a method of modulating an immune response comprising administering a derivatized pyridazine or a pharmaceutically acceptable salt thereof to a subject in need thereof.
  • a method of modulating an immune response in a subject comprising administering a compound of formula I:
  • R 1 and R 2 are lower alkyl, or (ii) together form a bridge in subformula I*
  • A, B, D, and E are, independently of one another, N or CH, with the stipulation that not more than 2 of these radicals are N;
  • G is lower alkylene, lower alkylene substituted by acyloxy or hydroxy, -CH 2 -O-, - CH 2 -S-, -CH 2 -NH-, oxa (-O-), thia (-S-), or imino (-NH-);
  • Q is lower alkyl, especially methyl;
  • R is H or lower alkyl;
  • X is imino, oxa, or thia;
  • Y is aryl, pyridyl, or unsubstituted or substituted cycloalkyl; and Z is mono- or disubstituted amino, halogen, alkyl, substituted alkyl,
  • G is methylene.
  • r is 0.
  • n is 0.
  • Y is substituted phenyl.
  • said phenyl is substituted with chloro.
  • said chloro is bound para to the phenyl group's point of attachment.
  • said modulating is inducing.
  • said inducing stimulates production of cytokines, chemokines, and/or growth factors.
  • said compound is administered in a subcytotoxic amount to said subject.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • the subject is not suffering from cancer.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels.
  • said compound has an average steady state drug concentration in the blood of less than 20 /xM.
  • subject is suffering from an autoimmune disease. Further still, said autoimmune disease is multiple sclerosis or Crohn's disease.
  • said subject is suffering from a viral infection.
  • said viral infection is HCV, HIV, or HSV.
  • said subject is suffering from allergies.
  • said subject is suffering from asthma.
  • the subject is suffering from precancerous lesions, such as actinic keratosis, atypical or dysplastic nevi, or premalignant lentigos.
  • precancerous lesions such as actinic keratosis, atypical or dysplastic nevi, or premalignant lentigos.
  • the subject is suffering from a disease associated with abnormal cellular proliferation, such as, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • the invention also relates to a compound of formula I, or a pharmaceutically acceptable salt thereof, or an N-oxide thereof, for use in modulating an immune response of a subject, wherein said compound n is 0 and any of r, m, R 1 , R 2 , A, B, D, E, G, Q, R, X 5 Y and Z is as defined above or below.
  • the invention also relates to a compound of the formula I, a salt thereof or an N-oxide thereof, wherein n is 0 and X is imino or thia, and any of r, m, R 1 , R 2 , A, B, D, E, G, Q, R, Y and Z is as defined above or below.
  • the prefix "lower” denotes a radical having up to and including a maximum of 7, especially up to and including a maximum of 4 carbon atoms, the radicals in question being either linear or branched with single or multiple branching.
  • the plural form is used for compounds, salts, and the like, this is taken to mean also a single compound, salt, or the like.
  • Any asymmetric carbon atoms may be present in the (R)-, (S)- or (R,S)-configuration, preferably in the (R)- or (S)- configuration.
  • Substituents at a double bond or a ring may be present in cis (-Z-) or trans (-E-) form.
  • the compounds may thus be present as mixtures of isomers or as pure isomers, preferably as enantiomer-pure diastereomers.
  • the radicals are as defined above for compounds of formula I.
  • ring members T 1 , T 2 , T 3 , and T 4 preferably only one is nitrogen, the remaining three being CH; preferably only T 3 , especially T 4 , is nitrogen, whereas the other ring members T 1 , T 2 , and T 4 or T 1 , T 2 , and T 3 are CH.
  • the index r is preferably 0 or 1.
  • the index n is preferably 0 or 1 , especially 0.
  • the index m is preferably 0, 1 , or 2, especially 0 or also 1.
  • ring members A, B, D, and E in formula I not more than 2 are N, and the remaining ones are CH.
  • each of the ring members A, B, D and E are CH.
  • G is a bivalent group -CH 2 -O-, -CH 2 -S-, or -CH 2 -NH-
  • the methylene group in each case is bound to the ring with ring members A, B, D r and E, whereas the heteroatom (O, S, or NH) is bound to the phthalazine ring in formula I.
  • Lower alkylene G may be branched or preferably linear and is especially branched or preferably linear Ci -C 4 alkylene, especially methylene (-CH 2 - ), ethylene (-CH 2 -CH 2 -), trimethylene (-CH 2 -CH 2 -CH 2 -) or tetramethylene (-CH 2 - CH 2 -CH 2 -CH 2 -). G is preferably methylene.
  • Acyl in lower alkylene substituted by acyloxy is preferably arylcarbonyloxy, wherein aryl is defined as below, especially benzoyloxy or lower alkanoyloxy, especially benzoyloxy; lower alkylene substituted by acyloxy is especially methylene substituted by benzoyloxy.
  • Lower alkylene substituted by hydroxy is preferably hydroxymethylene (-CH(OH)-).
  • G as lower alkylene substituted by acyloxy or hydroxy is preferred, or G as otherwise defined hereinbefore and hereinafter is in each case especially preferred.
  • Lower alkyl is especially Ci -C 4 alkyl, e.g. n-butyl, sec-butyl, tert- butyl, n-propyl, isopropyl, or especially methyl or also ethyl.
  • aryl is an aromatic radical having 6 to 14 carbon atoms, especially phenyl, naphthyl, fluorenyl or phenanthrenyl, the radicals defined above being unsubstituted or substituted by one or more, preferably up to three, especially one or two substituents, especially selected from amino, mono- or disubstituted amino, halogen, alkyl, substituted alkyl, hydroxy, etherified or esterified hydroxy, nitro, cyano, carboxy, esterified carboxy, alkanoyl, carbamoyl, N-mono- or N,N-disubstituted carbamoyl, amidino, guanidino, mercapto, sulfo, phenylthio, phenyl-lower alkylthio, alkylphenylthio, phenylsulfmyl, phenyl-lower alkylsulfmyl,
  • Mono- or disubstituted amino is especially amino substituted by one or two radicals selected independently of one another from lower alkyl, such as methyl; hydroxy-lower alkyl, such as 2-hydroxyethyl; phenyl-lower alkyl; lower alkanoyl, such as acetyl; benzoyl; substituted benzoyl, wherein the phenyl radical is unsubstituted or especially substituted by one or more, preferably one or two, substituents selected from nitro or amino, or also from halogen, amino, N-lower alkylamino, N,N-di-lower alkylamino, hydroxy, cyano, carboxy, lower alkoxycarbonyl, lower alkanoyl, and carbamoyl; and phenyl-lower alkoxycarbonyl, wherein the phenyl radical is unsubstituted or especially substituted by one or more, preferably one or two, substituents selected from
  • Halogen is especially fluorine, chlorine, bromine, or iodine, especially fluorine, chlorine, or bromine and halo is especially fluoro, chloro, bromo, or iodo, especially fluoro, chloro, or bromo.
  • alkyl has up to a maximum of 12 carbon atoms and is especially lower alkyl, especially methyl, or also ethyl, n- propyl, isopropyl, or tert-butyl.
  • Substituted alkyl is alkyl as last defined, especially lower alkyl, preferably methyl, where one or more, especially up to three, substituents may be present, primarily from the group selected from halogen, especially fluorine, and also from amino, N-lower alkylamino, N,N-di-lower alkylamino, N-lower alkanoylamino, hydroxy, cyano, carboxy, lower alkoxycarbonyl, and phenyl-lower alkoxycarbonyl. Trifluoromethyl is especially preferred.
  • Etherified hydroxy is especially C 8 -C 20 alkyloxy, such as n-decyloxy, lower alkoxy (preferred), such as methoxy, ethoxy, isopropyloxy, or n-pentyloxy, phenyl-lower alkoxy, such as benzyloxy, or also phenyloxy, or as an alternative or in addition to the previous group C 8 -C 20 alkyloxy, such as n-decyloxy, halogen-lower alkoxy, such as trifluoromethyloxy or 1,1,2,2-tetrafluoroethoxy.
  • C 8 -C 20 alkyloxy such as n-decyloxy, lower alkoxy (preferred), such as methoxy, ethoxy, isopropyloxy, or n-pentyloxy, phenyl-lower alkoxy, such as benzyloxy, or also phenyloxy, or as an alternative or in addition to the previous
  • Esterified hydroxy is especially lower alkanoyloxy, benzoyloxy, lower alkoxycarbonyloxy, such as tert-butoxycarbonyloxy, or phenyl-lower alkoxycarbonyloxy, such as benzyloxcarbonyloxy.
  • Esterified carboxy is especially lower alkoxycarbonyl, such as tert- butoxycarbonyl or ethoxycarbonyl, phenyl-lower alkoxycarbonyl, or phenyloxycarbonyl .
  • Alkanoyl is primarily alkylcarbonyl, especially lower alkanoyl, e.g. acetyl.
  • N-mono- or N,N-disubstituted carbamoyl is especially substituted by one or two substituents, lower alkyl, phenyl-lower alkyl, or hydroxy- lower alkyl, at the terminal nitrogen atom.
  • Alkylphenylthio is especially lower alkylphenylthio.
  • Alkylphenylsulfmyl is especially lower alkylphenylsulfmyl.
  • Alkylphenylsulfonyl is especially lower alkylphenylsulfonyl.
  • Pyridyl Y is preferably 3- or 4-pyridyl.
  • Unsubstituted or substituted cycloalkyl is preferably C 3 -C 8 cycloalkyl, which is unsubstituted or substituted in the same way as aryl, especially as defined for phenyl. Cyclohexyl or also cyclopentyl or cyclopropyl are preferred.
  • Heterocyclyl is especially a five or six-membered heterocyclic system with 1 or 2 heteroatoms selected from the group comprising nitrogen, oxygen, and sulfur, which may be unsaturated or wholly or partly saturated, and is unsubstituted or substituted especially by lower alkyl, such as methyl; a radical selected from 2- methylpyrimidm-4-yl, oxazol-5- yl, 2-methyl-l,3-dioxolan-2-yl, lH-pyrazol-3-yl, and l-methyl-pyrazol-3- yl is preferred.
  • Aryl in the form of phenyl which is substituted by lower alkylene dioxy bound to two adjacent C-atoms, such as methylenedioxy, is preferably 3,4- methylenedioxyphenyl .
  • the bonds in formula I characterized by wavy lines are present either as single or as double bonds. Preferably both are at the same time either single or double bonds.
  • An N-oxide of a compound of formula I is preferably an N-oxide in which a phthalazine-ring nitrogen or a nitrogen in the ring with ring members A, B, D, and E carries an oxygen atom, or several of the said nitrogen atoms carry an oxygen atom.
  • Salts are especially the pharmaceutically acceptable salts of compounds of formula I (or an N-oxide thereof).
  • Such salts are formed, for example, as acid addition salts, preferably with organic or inorganic acids, from compounds of formula I (or an N- oxide thereof with a basic nitrogen atom, especially the pharmaceutically acceptable salts.
  • Suitable inorganic acids are, for example, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid.
  • Suitable organic acids are, for example, carboxylic, phosphonic, sulfonic or sulfamic acids, for example acetic acid, propionic acid, octanoic acid, decanoic acid, dodecanoic acid, glycolic acid, lactic acid, 2- hydroxybutyric acid, gluconic acid, glucosemonocarboxylic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, glucaric acid, galactaric acid, amino acids, such as glutmic acid, asparic acid, N-methylglycine, acetylaminoacetic acid, N-acetylasparagine or N- acetylcysteine, pyruvic acid, acetoacetic acid, phosphoserine, 2 ⁇ or 3- glycerophosphoric acid, glucose-6-pliosphonc acid, glucose-
  • salts may also be formed with bases, e.g. metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethylamine or tri(2-hydroxyethyl)amine, or heterocyclic bases, for example N-ethylpipperidine or N, N'-dimethylpiperazine.
  • bases e.g. metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethylamine or tri(2-hydroxyethyl)amine, or heterocyclic bases, for example N-ethylpipperidine or N, N'-dimethylpiperazine.
  • a compound of formula I may also form internal salts.
  • pharmaceutically unacceptable salts for example picrates or perchlorates.
  • pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
  • any reference to the free compounds hereinbefore and hereinafter is to be understood as referring also to the corresponding salts, as appropriate and expedient.
  • the compounds of formula I (or an N-oxide thereof have valuable pharmacological properties, as described hereinbefore and hereinafter.
  • one of the ring members Ti, T 2 , T 3 and T 4 is nitrogen, and the others are in each case CH, and the binding is achieved via Ti and T 4 ;
  • A, B, D, and E are in each case CH, or also A, D, and E are each CH and B is N;
  • G is lower alkylene, especially methylene or ethylene (-CH 2 -CH 2 -), -CH 2 -NH-, -CH 2 - O-, hydroxymethylene, or benzoyloxymethylene, Q is methyl, which is bound to A, D, or to A and D;
  • R is H or lower alkyl, especially H or methyl,
  • X is imino, oxa, or thia
  • Y is phenyl, which is unsubstituted or is substituted by one or two substituents independently of one another from the group comprising amino; lower alkanoylamino, especially acetylamino; halogen, especially fluorine, chlorine
  • Preferred SMIPs in accordance with the fourth aspect of the invention are derivatized pyridazines, such as l-(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine, as well as analogs disclosed in US 6,258,812. More particularly, preferred SMIPs include derivatized pyridazines encompassed by Formula I as described in this section, or contained within the aforementioned references. [00272] Reference to "derivatized pyridazines" indicates compounds having the general structure of Formula I as described in this section.
  • a preferred derivatized pyridazine is a phthalazine such as l-(4-Chloroanilino)-4-(4- pyridylmethyl)phthalazine.
  • Ri and R 9 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a Rb, -C(O)Rc, -S(O) n Rd, or heterocyclyl;
  • R 3 and R 8 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a Rb, -C(O)Rc, -S(O) n R d , or heterocyclyl;
  • R 4 and R 7 are each independently H, -CN, -OH, halogen, alkyl, -0-(CH 2 ) q -R e , - C(O)Rc, -0-(CH 2 ) q -0-Re, -NR a R b , and -S(O) n Ra;
  • R 5 and R 6 are each independently H, -CN, -OH, halogen, al
  • Ri and R 9 are both H.
  • Rio is H.
  • R 3 is methyl.
  • r is 1 and R] 2 is H.
  • R 4 is -OCH 3 and R 5 is H.
  • R 4 is -CH 2 OH.
  • R 4 is -C(O)R 0 .
  • Another embodiment of the invention provides a method of modulating an immune response in a subject comprising administering a compound selected from the group consisting of:
  • said modulating is inducing.
  • said inducing stimulates production of cytokines, chemokines, and/or growth factors.
  • said compound is administered in a subcytotoxic amount to said subject.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels.
  • said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • subject is suffering from an autoimmune disease. Further still, said autoimmune disease is multiple sclerosis or Crohn's disease.
  • said subject is suffering from a viral infection.
  • said viral infection is HCV, HIV, or HSV.
  • said subject is suffering from allergies.
  • said subject is suffering from asthma.
  • the subject is suffering from a disease associated with abnormal cellular proliferation, such as, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • a disease associated with abnormal cellular proliferation such as, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • Any asymmetric carbon atoms may be present in the (R)-, (S)- or (R,S)-configuration, preferably in the (R)-
  • Substituents at a double bond or a ring may be present in cis (-Z-) or trans (-E-) form.
  • the compounds may thus be present as mixtures of isomers or as pure isomers, preferably as enantiomer-pure diastereomers.
  • Preferred SMIPs in accordance with the fifth aspect of the invention are staurosporine analogs, such as 9,13-Epoxy-lH,9H-diindolo[l,2,3-gh:3',2',r- lm]pyrrolo[3 ,4-j] [ 1 ,7]benzodiazonin- 1 -one, 2,3 , 10, 11 , 12, 13 -hexahydro- 11,12- dihydroxy- 10-methoxy-9-methyl; 9,13 -Epoxy- 1 H,9H-diindolo[ 1 ,2,3 -gh: 3 ',2', 1'- lm]pyrrolo[3,4-j][l,7]benzodiazonin-l-one, 2,3,10,11, 12,13-hexahydro-10-methoxy- ⁇ -methyl-ll- ⁇ ethylam ⁇ S-Epoxy-lH ⁇ H-diindolotl ⁇ -
  • preferred SMIPs include flavones, isoflavones and those encompassed by Formula I as described in this section, or contained within the aforementioned references.
  • Reference to "staurosporine analogs" indicates compounds having the general structure of Formula I as described in this section.
  • Preferred staurosporine analogs are selected from the group consisting of 9,13-Epoxy-lH,9H-diindolo[l,2,3- gh: 3',2', 1 '-lm]pyrrolo[3,4-j] [ 1 ,7]benzodiazonin- 1 -one, 2,3, 10, 11 , 12, 13 -hexahydro- 11,12-dihydroxy- 10-methoxy-9-methyl; 9, 13 -Epoxy- 1 H,9H-diindolo[ 1 ,2,3-gh: 3 ⁇ 2 ⁇ 1 '- lm]pyrrolo[3,4-j] [ 1 ,7]benzodiazonin- 1 -one, 2,3 , 10, 11 , 12, 13-hexahydro- 10-methoxy- 9-methyl-ll-(methylamino); 9,13-Epoxy-lH,9H-diindolo[l,2,3-gh:3',2',
  • One embodiment of the invention provides a method of treating a subject suffering from a viral infection comprising administering a small molecule in an amount such that said small molecule concomitantly inhibits viral replication while stimulating an immune response in said subject.
  • said small molecule is a nucleoside analog.
  • said small molecule inhibits viral DNA polymerase.
  • said small molecule is a nucleoside analog.
  • said small molecule is a substituted purine or pyrimidine.
  • Another embodiment of the invention provides a method of stimulating an immune response in a subject comprising administering a nucleoside analog of formula I:
  • R 3 is absent, H, Ci -6 alkyl, substituted Ci -6 alkyl, C 6- I 0 aryl, substituted C ⁇ -io aryl, heterocyclyl, or substituted heterocyclyl;
  • R 4 and R 5 are each independently H, halo, heterocyclyl, substituted heterocyclyl, -C(O)-R d , Ci- 6 alkyl, substituted Ci -6 alkyl, or bound together to form a 5 membered ring as in R 4-5 :
  • R4-5 the binding being achieved at the bonds indicated by a ⁇ ⁇ V ;
  • R 8 is H, halo, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, -OH, -NR a R b , -(CH 2 ) n -O-R c , -O- (Ci -6 alkyl), -S(O) p Re, or -C(O)-R 4 ;
  • Ri is -NH 2 .
  • each R f is H.
  • R 3 is -(CH 2 ) n -O-R c .
  • R 3 is -CH 2 -O-(C 2 alkyl or substituted C 2 alkyl)-O-(H, phosphate, diphosphate, triphosphate, Ci -6 alkyl or substituted Ci -6 alkyl).
  • R 3 is R 3a : R 3 a the binding being achieved at the bond indicated by a - ⁇ ;
  • R 6 and R 7 are each independently H, halo, Ci -6 alkoxy, substituted Ci -6 alkoxy, C 2-6 alkenyl, substituted C 2-6 alkenyl, -NR a Rb, -N 3 , or -OH;
  • R 6a and R 7a are each independently H, halo, or Ci -6 alkyl; each R a and R b is independently H, Ci -6 alkyl, substituted Ci -6 alkyl, -C(O)Rd, or C 6-I0 aryl;
  • each R d is independently H, halo, Q -6 alkyl, substituted Ci -6 alkyl, Ci -6 alkoxy, substituted Ci -6 alkoxy, -NH 2 , -NH(Ci -6 alkyl), -NH(substituted Ci -6 alkyl),
  • R 3 is R 3a
  • R 3 is R 3a
  • R 2 0.
  • R 4 and R 5 are both H.
  • Ri is -NH 2 .
  • Rf is H.
  • R 7 is H, F, or -OH.
  • R 3 is R 3a
  • R 6 is - N 3 .
  • R 3 is R 3a
  • R 6a and R 7a are both H.
  • R 331 In another embodiment wherein R 3 is R 3a , both R 7 and R 7a are methyl.
  • said substituted heterocyclyl within R 9 is Rc> a :
  • Rio and Rn are each independently H, halo, Ci -6 alkoxy, substituted Ci -6 alkoxy, - NR 3 Rb, or -OH;
  • R a and R b are independently H, Ci -6 alkyl, substituted Ci -6 alkyl, -C(O)R d , or C 6-1O aryl;
  • R d is H, halo, Ci -6 alkyl, substituted Ci -6 alkyl, Ci -6 alkoxy, substituted Ci -6 alkoxy, - NH 2 , -NH(Ci -6 alkyl), -NH(substituted Ci -6 alkyl), -N(Ci -6 alkyl) 2 , -N(substituted Ci -6 alkyl) 2 , C 6- Io aryl, or heterocyclyl; and Rf is independently H, C 1-6 alkyl, substituted Ci -6 alkyl, substituted Ci -6 alkyl, substituted Ci
  • R 8 is H, halo, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, heterocyclyl, substituted heterocyclyl, -OH, -NR a R b , -(CH 2 ) n -O-R c , -0-(C w alkyl), -S(O) p R e , or -C(O)-R d ;
  • R9 is H, Ci -6 alkyl, substituted Ci -6 alkyl, heterocyclyl, or R 9a :
  • Rio and Rn are each independently H, halo, Ci -6 alkoxy, substituted Ci -6 alkoxy, - NR a R b) or -OH; each R a and R b is independently H, C 1-6 alkyl, substituted Ci -6 alkyl, -C(O)Rd, or C 6- io aryl; each R 0 is independently H, phosphate, diphosphate, triphosphate, Ci -6 alkyl, substituted Ci -6 alkyl; each Ra is independently H, halo, Ci -6 alkyl, substituted C] -6 alkyl, Ci -6 alkoxy, substituted Ci -6 alkoxy, -NH 2 , -NH(Ci -6 alkyl), -NH(substituted Ci -6 alkyl), -N(Ci -6 alkyl) 2 , -N(substituted Ci -6 alkyl) 2 ,
  • R 9 is Rg a and R f is H.
  • Ri 0 and Rn are both H.
  • Ri 0 and Rn are both -OH.
  • Another embodiment of the invention provides a method of stimulating an immune response in a subject comprising administering a compound of formula III:
  • R 4 and R 5 are each independently H, halo, heterocyclyl, substituted heterocyclyl, -C(O)-Rd, Ci -6 alkyl, or substituted Ci -6 alkyl;
  • R 6 and R 7 are each independently H, halo, Ci -6 alkoxy, substituted Ci -6 alkoxy, C 2-6 alkenyl, substituted C 2-6 alkenyl, -NR 2 R b , -N3, or -OH;
  • each Ra and R b is independently H, Ci -6 alkyl, substituted Ci -6 alkyl, -C(O)R d , or C 6
  • composition comprising a cytokine or chemokine in combination with any compound(s) disclosed herein, such as a nucleoside, a compound encompassed by formula I, II, or III, or those listed in Table 1.
  • a nucleoside a compound encompassed by formula I, II, or III, or those listed in Table 1.
  • the compound is present in the blood at less then 2O uM.
  • said nucleoside analog induces interferon biosynthesis.
  • said nucleoside analog is co-administered with another agent to the subject.
  • the agent is an antigen.
  • the agent is a vaccine.
  • nucleoside analog induces the production of TNF-cein the subject.
  • the nucleoside analog has an average steady-state drug concentration in the blood of less than 20 ⁇ M.
  • the subject is suffering from a microbial infection.
  • the subject is suffering from a viral infection.
  • the viral infection is a viral infection caused by the hepatitis C virus (HCV).
  • the viral infection is caused by the human immunodeficiency virus (HIV).
  • the viral infection is caused by the herpes simplex virus (HSV).
  • the viral infection is caused by the SARS virus.
  • any asymmetric carbon atoms for example in compounds of formula I [or an N-oxide thereof]) in the (R)-, (S)- or (Re ⁇ configuration, preferably in the (R)- or (S)- configuration.
  • Substituents at a double bond or a ring may be present in cis (-Z-) or trans (-E-) form.
  • the compounds may thus be present as mixtures of isomers or as pure isomers, preferably as enantiomerically-pure.
  • the invention encompasses all possible combinations of the preceding embodiments.
  • the nucleoside analog compounds can be used with or without an antigen in therapeutic applications, for example to treat cancer or infectious diseases.
  • the nucleoside analog compounds also may be used in combination with other therapeutic agents, such as anti-virals and monoclonal antibodies in different therapeutic applications.
  • One preferred embodiment of the method of inducing an immunostimulatory effect in a patient is directed to administering an immunogenic composition comprising a vaccine in an amount effective to stimulate an immune response such as a cell-mediated immune response and, as a vac ⁇ ine adjuvant, an nucleoside analog compound, in an amount effective to potentiate the immune response such as the cell-mediated immune response to the vaccine.
  • Agents combined with the nucleoside analog compounds, contemplated to be useful in treating the aforementioned diseases include those well known in the art, such as, anesthetics, hypnotic sedatives, anti-anxieties, antiepileptics, antipyretic antiphlogistics, stimulants, wake amines, anti-parkinson drugs, agents for psychoneuroses, agents for central nervous system, skeletal muscle relaxants, agents for autonomic nervous system, antispastic agents, cytotoxic agents, monoclonal antibodies, drugs for eye, drugs for nose and ear, anti-vertiginous drugs, cardiotonics, antiarrhythmic drugs, diuretics, pressure reduction drugs, vasoconstrictors, coronary vaso-dilators, peripheral vasodilating drugs, hyper-lipemia drugs, breath stimulants, antitussive and expectorant drugs, bronchodilators, drugs for allergy, antidiarrheal drugs, drugs for intestinal disorders, peptic ulcer drugs,
  • methods of treating allergies comprising administering an nucleoside analog compound alone or in combination with another agent known to be effective against allergies, wherein said combination is more effective in treating an allergic condition than the known agent(s) are without the addition of said nucleoside analog compound.
  • the known agent is an antihistamine and/or a leukotriene inhibitor.
  • the allergic condition is asthma.
  • the allergic condition is selected from the group consisting of allergic rhinitis, dermatosis, and urticaria.
  • the combination is administered to a subject enterally, parenterally, intranasally, subcutaneously, or intraarterially.
  • Preferred SMIPs in accordance with the sixth aspect of the invention include nucleoside analogs and those compounds encompassed by Formula I as described in this section, or contained within any reference cited herein.
  • a "substituted pyrimidine” refers to an aromatic ring containing two nitrogen
  • a "substituted purine” refers to two condensed aromatic rings containing two
  • One embodiment provides a method of modulating an immune response in a subject comprising administering a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate.
  • a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezom
  • Another embodiment is provided wherein said compound is co ⁇ administered with another agent.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • the subject is not suffering from cancer.
  • said subject is suffering from a cancer selected from the group consisting of prostate, breast, ovarian, colon, epidermal, ductal, non-small-cell lung, colorectal, neuroendocrine, spinal, esophageal, pancreas, renal, stomach, lymphoidal, intestinal, bladder, uterine cervix, head and neck, brain, nasopharynx, leukemia, kaposis sarcoma, and mesothelioma.
  • a cancer selected from the group consisting of prostate, breast, ovarian, colon, epidermal, ductal, non-small-cell lung, colorectal, neuroendocrine, spinal, esophageal, pancreas, renal, stomach, lymphoidal, intestinal, bladder, uterine cervix, head and neck, brain, nasopharynx, leukemia, kaposis sarcoma, and mesothelioma.
  • Another embodiment is provided wherein said compound is co-
  • One embodiment provides a method of stimulating an immune response in a subject comprising administering a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-Ol, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate.
  • said compound is co ⁇ administered with another agent.
  • said compound is administered in a dose capable of increasing TNF-cn levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • said compound is administered in a dose capable of inducing cytokines.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • the subject is not suffering from cancer.
  • said subject is suffering from a cancer selected from the group consisting of prostate, breast, ovarian, colon, epidermal, ductal, non-small-cell lung, colorectal, neuroendocrine, spinal, esophageal, pancreas, renal, stomach, lymphoidal, intestinal, bladder, uterine cervix, head and neck, brain, nasopharynx, leukemia, kaposis sarcoma, and mesothelioma.
  • a cancer selected from the group consisting of prostate, breast, ovarian, colon, epidermal, ductal, non-small-cell lung, colorectal, neuroendocrine, spinal, esophageal, pancreas, renal, stomach, lymphoidal, intestinal, bladder, uterine cervix, head and neck, brain, nasopharynx, leukemia, kaposis sarcoma, and mesothelioma.
  • Another embodiment is provided wherein said compound is co-
  • Another embodiment of the invention provides a method of treating a subject in need of immune stimulation, comprising administering to a subject in need thereof, a subcytotoxic amount of a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 8
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • said amount is sufficient to stimulate cytokine production in the subject.
  • said subject is in remission from cancer.
  • said compound is administered for the treatment of refractory cancer cells.
  • said compound is administered metronomically.
  • the subject is not suffering from cancer.
  • said subject is suffering from a cancer selected from the group consisting of prostate, breast, ovarian, colon, epidermal, ductal, non-small-cell lung, colorectal, neuroendocrine, spinal, esophageal, pancreas, renal, stomach, lymphoidal, intestinal, bladder, uterine cervix, head and neck, brain, nasopharynx, leukemia, kaposis sarcoma, and mesothelioma.
  • a cancer selected from the group consisting of prostate, breast, ovarian, colon, epidermal, ductal, non-small-cell lung, colorectal, neuroendocrine, spinal, esophageal, pancreas, renal, stomach, lymphoidal, intestinal, bladder, uterine cervix, head and neck, brain, nasopharynx, leukemia, kaposis sarcoma, and mesothelioma.
  • Another embodiment is provided wherein said compound is co-
  • Another embodiment of the invention provides a method of identifying a subject in need of a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP- 701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY- 317615, squalamine, UCN-Ol, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate,
  • a further step of administering said compound to the subject, wherein upon administration of said compound, cytokine levels increase is provided.
  • Another embodiment is provided wherein said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • Another embodiment of the invention provides a method of identifying a subject in need of a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP- 701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY- 317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate, comprising: a) taking a blood sample from said subject; b) monitoring for leukocyte levels in said sample; and c) identifying said subject by decreased leukocyte levels in said sample.
  • a further step of administering said compound to the subject, wherein upon administration of said compound, leukocyte levels increase is provided.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • Another embodiment of the invention provides a method of determining efficacy of a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP- 701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotmib, perifosine, CYC-202, LY- 317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate, comprising: a) taking a first blood sample from a subject; b) identifying a first cytokine level in said sample; c) administering said compound to said subject; d) taking a second
  • Another embodiment of the invention provides a method of treating a subject suffering from an autoimmune disease, comprising administering to a subject in need thereof, a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP- 701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY- 317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate.
  • said autoimmune disease is Addison's Disease, or Alopecia Areata, or Ankylosing Spondylitis, or Antiphospholipid Syndrome (APS), or Behcet's Disease, or Chronic Fatigue Syndrome, or Crohn's Disease, or Ulcerative Colitis, or Diabetes, or Fibromyalgia, or Goodpasture Syndrome, or Graft Versus Host Disease, or Graves' Disease, or Guillain-Barre Syndrome, or Lupus, or Meniere's, or Multiple Sclerosis, or Myasthenia Gravis, or Myositis, or Pemphigus Vulgaris, or Primary Biliary Cirrhosis, or Psoriasis, or Rheumatic Fever, or Sarcoidosis, or Scleroderma, or Vasculitis, or Vitiligo, or Wegener's Granulomatosis.
  • APS Antiphospholipid Syndrome
  • Behcet's Disease or Chronic Fatigue Syndrome, or Crohn's Disease, or Ulcerative Colitis
  • Diabetes or Fibro
  • Another embodiment is provided wherein said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF-a levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • Another embodiment of the invention provides a method of treating a subject suffering from a viral infection, comprising administering to a subject in need thereof, a compound selected from the group consisting of fenretinide, vatalanib, SU- 11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate.
  • said viral infection is HTV, or HCV, or HBV, or HSV.
  • said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF-ce levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • Another embodiment of the invention provides a method of treating a subject suffering from allergies or asthma, comprising administering to a subject in need thereof, a compound selected from the group consisting of fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD- 6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate.
  • Another embodiment is provided wherein said compound is co-administered with another agent.
  • said compound is administered in a dose capable of increasing TNF- ⁇ levels, more specifically, said compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • Preferred SMIPs in accordance with the seventh aspect of the invention include fenretinide, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortezomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib, erlotinib, perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate compositions as well as analogs disclosed in the following patents and patent applications: US 4,323,581, US 6,258,812, WO 98/35958, WO 01/60814, US 5,883,113, WO 99/61422, US 5,883,113,
  • preferred SMIPs include those encompassed by Formula I in the aforementioned patents and patent applications.
  • the invention also includes compounds encompassed by Formula I, disclosed in the following patents and patent applications: US 4,323,581, US 6,258,812, WO 98/35958, WO 01/60814, US 5,883,113, WO 99/61422, US 5,883,113, WO 99/61422, WO 03/24978, WO 03/04505, US 5,780,454, US 2003134846, WO 97/21701, US 5,621,100, WO 01/32651, WO 02/16351, US 6,727,256, WO 02/02552, US 5,457,105, US 5,616,582, US 5,770,599, US 5,747,498, WO, 96/30347, US 2003171303, WO 97/20842, WO 99/02162, WO 95/17182, WO 01/79255, WO 89/07105, US 5,439,936,
  • IC50 value The concentration of an inhibitor that causes a 50 % reduction in a measured activity.
  • IFN Interferon IL Interleukin IMS Immunomagnetic separation IPV Inactivated polio virus LCMS Liquid Chromatography / Mass Spectroscopy LPS Lip polysaccharide MAb or mAb Monoclonal Antibody Men A Neisseria Meningitidis Type A Men C Neisseria Meningitidis Type C
  • SIP small molecule immunopotentiating compounds
  • small molecule compounds below about MW 800 g/mol, capable of stimulating or modulating a pro-inflammatory response in a patient.
  • the SMIP compounds are able to stimulate human peripheral blood
  • SMIS refers to small molecule immunosuppressant compounds, including small molecule compounds below about MW 800 g/mol, capable of suppressing or modulating an immune response in a patient.
  • the SMIS compounds are able to inhibit human peripheral blood
  • cytokines mononuclear cell's ability to produce cytokines, chemokines, and/or growth factors.
  • the SMIS compounds are able to induce TGF-beta
  • refractory cancer cells refers to cancer cell lines that are resistant to preexisting therapeutics or treatment regimens, including prescribed
  • allergen refers to a substance (antigen) that can induce an allergic or asthmatic response in a susceptible subject.
  • the list of allergens is enormous and can include pollens, insect venoms, animal dander, dust, fungal spores, and drugs (e.g. penicillin).
  • allergens refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively associated with atopic or allergic symptoms.
  • leukotriene inhibitor includes any agent or compound that inhibits, restrains, retards or otherwise interacts with the action or activity of leukotrienes, such as, but not limited to, 5 -lipoxygenase (“5-LO”) inhibitors, 5- lipoxygenase activating protein (“FLAP”) antagonists, and leukotriene D4 (“LTD4 ”) antagonists.
  • 5-LO 5 -lipoxygenase
  • FLAP 5- lipoxygenase activating protein
  • LTD4 leukotriene D4
  • Immuno-stimulation or “immune potentiation” refers to activation of the immune system, including humoral or cellular activation, for example, activation of a cell, such as a killer (T or NK) or dendritic cell of the immune system, for example, causing the increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • T or NK killer
  • Modulating an immune response refers to either immune potentiation or immune suppression as defined herein.
  • An "immunogenic composition” refers to a composition capable of modulating the production of cytokines in a subject thereby effecting immune potentiation in the subject.
  • Immuno suppression refers to deactivation of the immune system, for example, preventing or lessening cytokine production from a dendritic cell leading to an overall attenuation of host defense (immune response).
  • An “immune-stimulatory effective amount” is an amount effective for activation of the immune system, for example, causing the increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • Enhancing the immune response to an antigen by a compound refers to enhancement of the immune response in comparison to that in the absence of the compound.
  • An enhanced immune-response eliciting composition is a composition generally comprising an antigen and a small molecule immune potentiator compound that elicits an immune response greater that a composition comprising an antigen and not containing one or more small molecule immune potentiator compounds.
  • the compound acts as an adjuvant, for example for use in vaccine compositions and methods.
  • a "disease associated with cellular proliferation” includes, but is not limited to neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, and endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • the term "effective amount” is an amount necessary or sufficient to realize a desired biological effect.
  • an effective amount of a compound to treat an infectious disorder may be an amount necessary to cause an antigen specific immune response upon exposure to an infectious agent. The effective amount may vary, depending, for example, upon the condition treated, weight of the subject and severity of the disease.
  • an effective amount for treatment refers to an amount sufficient to palliate, ameliorate, stabilize, reverse, slow or delay progression of a condition such as a disease state.
  • Reference to "metronomic administration” or “administered metronomically” refers to increasingly frequent dosing regimens, at lower drug concentrations, as compared with known dosing regimens for an existing therapeutic. Metronomic administration varies from the typical dosing of cytotoxic drugs, which involves episodic (less frequent) administration at maximum tolerated doses (MTDs).
  • a "subject” or “patient” is meant to describe a human or vertebrate animal including a dog, cat, pocket pet, marmoset, horse, cow, pig, sheep, goat, elephant, giraffe, chicken, lion, monkey, owl, rat, squirrel, slender loris, and mouse.
  • a "pocket pet” refers to a group of vertebrate animals capable of fitting into a commodious coat pocket such as, for example, hamsters, chinchillas, ferrets, rats, guinea pigs, gerbils, rabbits and sugar gliders.
  • ester refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms.
  • Representative examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
  • the compounds of the present invention can be used in the form of salts as in "pharmaceutically acceptable salts" derived from inorganic or organic acids.
  • These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-napth- alenesulfonate, oxalate, pamoate
  • the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides such
  • prodrugs refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the formula as described herein, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S.
  • alkyl refers to substituted and unsubstituted alky groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • the phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -CH(CH 3 ) 2 , -CH(CH 3 )(CH 2 CH 3 ), -CH(CH 2 CH 3 ) 2 , -C(CH 3 ) 3 , -C(CH 2 CH 3 ) 3 , -CH 2 CH(CH 3 ) 2 , -CH 2 CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH(CH 2 CH 3 ) 2 , -CH 2 C(CH 3 ) 3 , -CH 2 C(CH 2 CHs) 3 , -CH(CH 3 )CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH 2 CH(CH 3 ) 2 , -CH 2 CH 2 CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH 2 CH(CH 3 ) 2 , -CH 2 CH(CH 3 )(CH 2 CH
  • the phrase also includes cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • alkyl also includes groups in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen and non-carbon atoms such as, but not limited to, a halogen atom in halides such as F, Cl, Br, and I; and oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as in trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups
  • Alkyl groups are those limited to having 1 to 20 carbon atoms and as many as 5 additional heteroatoms as described above. More preferred alkyl groups have from 1 to 5 carbon atoms and as many as 2 heteroatoms.
  • the term "Ci -6 alkyl” has the same meaning as alkyl, except that it is limited to alkyl groups of six carbon atoms or less.
  • the term “C 2 alkyl” indicates an alkyl group having two carbon atoms.
  • aryl refers to substituted and unsubstituted aryl groups that do not contain heteroatoms. Thus the phrase includes, but is not limited to, groups such as phenyl, biphenyl, anthracenyl, naphthenyl by way of example.
  • Aryl groups also include those in which one of the aromatic carbons is bonded to a non-carbon or non- hydrogen atoms described above (in the alkyl definition) and also includes aryl groups in which one or more aromatic carbons of the aryl group is bonded to a substituted and/or unsubstituted alkyl, alkenyl, or alkynyl group as defined herein.
  • aryl includes, but is not limited to tolyl, and hydroxyphenyl among others.
  • C 6- Io aryl has the same meaning as aryl, except that it is limited to aryl groups of from six to ten carbon atoms.
  • alkenyl refers to straight and branched chain and cyclic groups such as those described with respect to alkyl groups as defined above, except that at least one double bond exists between two carbon atoms.
  • Alkenyl groups are those limited to having 2 to 15 carbon atoms and as many as 4 additional heteroatoms as described above. More preferred alkenyl groups have from 2 to 5 carbon atoms and as many as 2 heteroatoms.
  • C 2-6 alkenyl has the same meaning as alkenyl, except that it is limited to alkenyl groups of from two to six carbon atoms.
  • alkoxy refers to substituted or unsubstituted alkoxy groups of the formula -O-alkyl, wherein the point of attachment is the oxy group and the alkyl group is as defined above.
  • Alkoxy groups are those limited to having 1 to 20 carbon atoms and as many as 5 additional heteroatoms, including the oxygen atom. More preferred alkoxy groups have from 1 to 5 carbon atoms and as many as 2 heteroatoms, including the oxygen atom.
  • the term "Ci -6 alkoxy” has the same meaning as alkoxy, except that it is limited to alkoxy groups of six carbon atoms or less.
  • alkynyl groups in which a non-carbon or non-hydrogen atom is bonded to a carbon triple bonded to another carbon and those in which a non-carbon or non-hydrogen atom is bonded to a carbon not involved in a triple bond to another carbon.
  • Alkynyl groups are those limited to having 2 to 15 carbon atoms and as many as 4 additional heteroatoms as described above. More preferred alkynyl groups have from 2 to 5 carbon atoms and as many as 2 heteroatoms.
  • the term "C 2-6 alkynyl" has the same meaning as alkynyl, except that it is limited to alkynyl groups of from two to six carbon atoms.
  • aryloxy refers to groups having the formula -O-aryl, wherein the point of attachment is the oxy group and the aryl group is as defined above.
  • C 6-1O aryloxy has the same meaning as aryloxy, except that it is limited to aryloxy groups of six to ten carbon atoms.
  • trihalomethyl refers to a methyl group in which the three H atoms of the methyl group are substituted with three halogens which may be same or different.
  • One example of such a group is a -CF 3 group in which all three H atoms of the methyl group are substituted with F atoms.
  • C 1-6 alkoxy-Ci- 6 alkyl refers to ether groups with as many as 12 carbon atoms.
  • Ci -6 alkoxy-Ci -6 alkyl group is -CH 2 -O-CH 2 CH 3 .
  • C 6-1O aryloxy-Ci -6 alkyl refers to aryl ether groups of 16 carbon atoms or less, especially of 10 carbon atoms or less bound at the Ci -6 alkyl group.
  • a C 6-1 O aryloxy-Ci -6 alkyl group is propoxybenzene.
  • C 6-1 O aryl-Ci -6 alkyl refers to arylalkyl groups of 16 carbon atoms or less, especially of 10 carbon atoms or less bound at the Ci -6 alkyl group.
  • C 6- io aryl-C ⁇ alkyl group is toluene.
  • heterocyclyl refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S.
  • heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g. 4H-l,2,4-triazolyl, lH-l,2,3-triazolyl, 2H-l,2,3-triazolyl etc.), tetrazolyl, (e.g.
  • saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms such as, but not limited to furanyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, oxazolyl, isoxazolyl, oxadiazolyl (e.g.
  • unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g.
  • 1,3-benzodioxoyl, etc. unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing an oxygen atom and 1 to 2 oxygen atoms such as benzoxathiinyl.
  • unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl
  • saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane
  • Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones).
  • heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1- dioxide.
  • Preferred heterocyclyl groups contain 5 or 6 ring members.
  • More preferred heterocyclyl groups include morphorine, piperazine, piperidine, pyrrolidine, imidazole, pyrazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, thiomorpholine, thiomorpholine in which the S atom of the thiomorpholine is bonded to one or more O atoms, pyrrole, homopiperazine, oxazolidin-2-one, pyrrolidin-2-one, oxazole, quinuclidine, thiazole, isoxazole, furan, and tetrahydrofuran.
  • Heterocyclyl also refers to those groups as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups (also referred to herein as “substituted heterocyclyl”). Examples, include, but are not limited to, 2-methylbenzimidazolyl, 5- methylbenzimidazolyl, 5-chlorobenzthiazolyl, 1 -methyl piperazinyl, and 2- chloropyridyl among others. Heterocyclyl groups are those limited to having 2 to 15 carbon atoms and as many as 6 additional heteroatoms as described above.
  • More preferred heterocyclyl groups have from 3 to 5 carbon atoms and as many as 2 heteroatoms.
  • substituted as applied to an undefined, yet well known in the art group, such as phenyl, will have the same meaning with respect to the optional appendages as described in the definition of alkyl.
  • substitution groups include, for example, hydroxyl, nitro, amino, imino, cyano, halo, thio, thioamido, amidino, imidino, oxo, oxamidino, methoxamidino, imidino, guanidino, sulfonamide, carboxyl, formyl, alkyl, heterocyclyl, aryl, haloalkyl, alkoxy, alkoxyalkyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkylthio, aminoalkyl, alkylamino, cyanoalkyl, phosphate, diphosphate, triphosphate and the like.
  • Ci -6 alkyl is tert-butanol.
  • the substitution group can itself be substituted one time.
  • an alkoxy substituent of an alkyl group may be substituted with a halogen, and oxo group, an aryl group, or the like.
  • the group substituted onto the substitution group can be carboxyl, halo, nitro, oxo, amino, cyano, hydroxyl, Ci -6 alkyl, C) -6 alkoxy, C 6 .
  • substituted substituent includes a straight chain group
  • substitution can occur either within the chain (e.g., 2-hydroxypropyl, 2-aminobutyl, and the like) or at the chain terminus (e.g., 2-hydroxyethyl, 3-cyanopropyl, and the like).
  • Substituted substituents can be straight chain, branched or cyclic arrangements of covalently bonded carbon atoms or heteroatoms.
  • phosphate indicates -O-PO 3 , where the point of attachment is oxo.
  • a "phosphate” group is attached at an oxo moiety (e.g. CH 3 -O- ⁇ hosphate) the phosphate substituent is -PO 3 (e.g. CH 3 -O-PO 3 ).
  • "Diphosphate” and “triphosphate” groups are respectively 2 and 3 phosphate moieties bound together as in -0-P(O) 2 -O-P(O) 2 -O-PO 3 for triphophate.
  • protected or a "protecting group” with respect to hydroxyl groups, amine groups, and sulfhydryl groups refers to forms of these functionalities which are protected from undesirable reaction with a protecting group known to those skilled in the art such as those set forth in Protective Groups in Organic Synthesis, Greene, T.W., John Wiley & Sons, New York, NY, (1st Edition, 1981) which can be added or removed using the procedures set forth therein.
  • Examples of protected hydroxyl groups include, but are not limited to, silyl ethers such as those obtained by reaction of a hydroxyl group with a reagent such as, but not limited to, t- butyldimethyl-chlorosilane, trimethylchlorosilane, triisopropylchlorosilane, triethylchlorosilane; substituted methyl and ethyl ethers such as, but not limited to methoxymethyl ether, methythiomethyl ether, benzyloxymethyl ether, t-butoxymethyl ether, 2-methoxyethoxymethyl ether, tetrahydropyranyl ethers, 1-ethoxyethyl ether, allyl ether, benzyl ether; esters such as, but not limited to, benzoylformate, formate, acetate, trichloroacetate, and trifiuoracetate.
  • a reagent such as, but not
  • protected amine groups include, but are not limited to, benzyl or dibenzyl, amides such as, formamide, acetamide, trifluoroacetamide, and benzamide; imides, such as phthalimide, and dithiosuccinimide; and others.
  • a protecting group for amines is a benzyl group.
  • protected sulfhydryl groups include, but are not limited to, thioethers such as S-benzyl thioether, and S-4-picolyl thioether; substituted S-methyl derivatives such as hemithio, dithio and aminothio acetals; and others.
  • the invention also includes isotopically-labeled compounds, that are structurally identical to those disclosed above, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F and 36 Cl, respectively.
  • Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds and of said prodrugs that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention.
  • Certain isotopically labeled compounds of the present invention, for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out known or referenced procedures and by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • Preferred "cytokines" include IL 1-30 as well as TNF-alpha, TNF-beta, IFN-alpha (family), IFN-beta and IFN-gamma.
  • IL 1-30 indicates interleukin cytokines selected from the group consisting of ILIA, ILlB, IL1F5, IL1F6, IL1F7, IL1F8, IL1F9, ILlFlO, ILlRl, IL1R2, ILlRAP, ILlRAPLl, IL1RAPL2, ILlRLl, IL1RL2, ILlRN 3 IL2, IL2RA, IL2RB, IL2RG, IL3, IL3RA, IL4, IL4R, IL5, IL5RA, IL6, IL6R, IL6RL1, IL6ST, IL6ST2, IL6STP, IL7, IL7R, IL8, IL8RA, IL8RB, IL8RBP, IL9, IL9R, IL9RP1, IL9RP2, IL9RP3, IL9RP4, ILlO, ILlORA
  • chemokines include IL-Ib, IL-2, IL-4, IL-5, IL-6, IL- 10, IL-12, and IL-13.
  • Reference to "chemokines” indicates: CXC chemokines including CXCLl 3 CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, CXCLlO, CXCLl 1, CXCL12, CXCL13, CXCL14, CXCL15, and CXCL16; C chemokines including XCLl, and XCL2; CX 3 C chemokines including CX 3 CLl; and CC chemokines including CCLl 3 CCL2, CCL3, CCL3L1, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9/CCL10, CCLI l, CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL
  • Vaccine compositions contemplated to be within the scope of the several aspects of the present invention may include (an) additional adjuvant(s).
  • Preferred adjuvants to enhance effectiveness of the composition include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc; (2) oil-in-water emulsion formulations (with or without specific immunostimulating agents such as muramyl peptides or bacterial cell wall components), such as, for example (a) MF59TM (WO90/14837), containing 5% squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing MTP-PE) formulated into submicron particles using a microfluidizer, (b) SAF, containing 5% squalene, 0.5% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RibiTM adjuvant system (RAS), (Ribi Immunochem,
  • cytokines such as interleukins (e.g. IL-I, IL-2, IL-4, IL- 5, IL-6, IL-7, IL- 12 (WO99/44636), etc.), interferons (e.g. gamma interferon), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), etc.; (6) momophosphoryl lipid A (MPL) or 3-0-deacylated MPL (3dMPL), optionally in the substantial absence of alum when used with pneumococcal saccharides e.g.
  • CFA Complete Freund's Adjuvant
  • IFA Incomplete Freund's Adjuvant
  • cytokines such as interleukins (e.g. IL-I, IL-2, IL-4, IL- 5, IL-6, IL-7, IL- 12 (WO99/44636), etc.), interferons (e.g. gamma interferon), macrophage colony stimulating factor
  • WO00/56358; and RC529 (7) combinations of 3dMPL with, for example, QS21 and /or oil-in-water emulsions e.g. EP-A-0835318; (8) oligonucleotides comprising CpG motifs, i.e. containing at least one CG dinucleotide, with 5-methylcytosine optionally being used in place of cytosine; (9) a polyoxyethylene ether or a polyoxyethylene ester e.g.
  • WO99/52549 (10) a polyoxyethylene sorbitan ester surfactant in combination with an octoxynol (WOO 121207) or a polyoxyethylene alkyl ether or ester surfactant in combination with at least one additional non-ionic surfactant such as an octoxynol (WO01/21152); (11) a saponin and an immunostimulatory oligonucleotide (e.g. a CpG oligonucleotide) (WO00/62800); (12) an immunostimulant and a particle of metal salt e.g WO00/23105; (13) a saponin and an oil-in-water emulsion e.g.
  • the invention is also directed to methods of administering the vaccine composition.
  • the vaccine is administered in an amount effective to stimulate an immune response.
  • the amount that constitutes an effective amount depends, inter alia, on the particular vaccine used, the particular adjuvant compound being administered and the amount thereof, the immune response that is to be enhanced (humoral or cell mediated), the state of the immune system (e.g., suppressed, compromised, stimulated), and the desired therapeutic result. Accordingly it is not practical to set forth generally the amount that constitutes an effective amount of the vaccine. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • the vaccine compositions of the invention can be administered to animals, e.g., mammals human and non-human, including, for example, pocket pets, fowl, and the like according to conventional methods well known to those skilled in the art (e.g., orally, subcutaneously, nasally, topically).
  • Suitable vaccines include, but are not limited to, any material that raises either humoral or cell mediated immune response, or both.
  • Suitable vaccines include live viral and bacterial antigens and inactivated viral, tumor-derived, protozoal, organism-derived, fungal, and bacterial antigens, toxoids, toxins, polysaccharides, proteins, glycoproteins, peptides, and the like.
  • vaccines such as those used in connection with BCG (live bacteria), cholera, plague, and typhoid (killed bacteria), hepatitis B, influenza, inactivated polio, and rabies (inactivated virus), measles, mumps, rubella, oral polio, SARS vaccines, and yellow fever (live virus), tetanus and diphtheria (toxoids), hemophilus influenzae b, meningococcal, and pneumococcal (bacterial polysaccharides) also can be used. Any antigen known in the art or disclosed herein may be used.
  • Exemplary experimental subunit antigens include those related to viral disease such as adenovirus, AIDS, chicken pox, cytomegalovirus, dengue, feline leukemia, fowl plague, hepatitis A, hepatitis B, hepatitis C, HSV-I, HSV-2, hog cholera, influenza A, influenza B, Japanese encephalitis, measles, parainfluenza, rabies, respiratory syncytial virus, SARS virus, rotavirus, wart, and yellow fever.
  • Specific antigens include: a protein antigen from N. meningitides serogroup B (1-7); an outer-membrane vesicle (OMV) preparation from N.
  • OMV outer-membrane vesicle
  • meningitides serogroup B (8, 9, 10, 11); a saccharide antigen from N. meningitides serogroup A, C W135 and/or Y, such as the oligosaccharide (12) from serogroup C (13); a saccharide antigen horn Streptococcus pneumoniae (14, 15, 16); an antigen from N.
  • gonorrhoeae (1, 2, 3); an antigen from Chlamydia pneumoniae (17, 18, 19, 20, 21, 22, 23); an antigen from Chlamydia trachomatis (24); an antigen from hepatitis A virus, such as inactivated virus (25, 26); an antigen from hepatitis B virus, such as the surface and/or core antigens (e.g. 26, 27); an antigen from hepatitis C virus (28); an antigen from Bordetella pertussis, such as petussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B.
  • Bordetella pertussis such as petussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B.
  • pertussis optionally also combination with pertactin and/or agglutinogens 2 and 3 (29, 30); a diphtheria antigen, such as a diphtheria toxoid (31:chapter 3) e.g.
  • a tetanus antigen such as a tetanus toxoid (31 : chapter 4); a protein antigen from Helicobacter pylori such as CagA (33), VacA (33), NAP (34), HopX (5), HopY (35) and/or urease; a saccharide antigen from Haemophilus influenzae B (13); an antigen from Porphyromonas gingivalis (36); polio antigen(s) (37, 38) such as IPV or OPV; rabies antigen(s) (39) such lyophilized inactivated virus (40, RabAvertTM); measles, mumps and/or rubella antigens (31: chapters 9, 10, & 11); influenza antigen(s) (31:chapter 19), such as the haemagglutinin and/or neuraminidase surface proteins; an antigen from Moraxella catarrhalis (41); an antigen from Moraxella catarr
  • the composition may comprise one or more of the above antigens.
  • a saccharide or carbohydrate antigen is used, it is preferably conjugated to a carrier protein in order to enhance antigenicity (47-56).
  • Preferred carrier proteins are bacterial toxins or toxoids, such as diphtheria or tetanus toxoids.
  • the CRM 1 C 17 diphtheria toxoid is particularly preferred.
  • Other suitable carrier proteins include the N. meningitides outer membrane protein (57), synthetic peptides (58, 59), heat shock proteins (60), pertussis proteins (61, 62), protein D from H. influenzae (63), toxin A or B from C. difficile (64) etc.
  • a mixture comprises capsular saccharides from both serogroups A and C
  • the ratio (w/w) of MenA saccharide:MenC saccharide is greater than 1 (e.g. 2:1, 3:1, 4:4, 5:1, 10:1 or higher).
  • Saccharides from different serogroups of ⁇ . meningitides may be conjugated to the same or different carrier proteins. [00434] Any suitable conjugation reaction can be used, with any suitable linker where necessary.
  • Toxic protein antigens may be detoxified where necessary (e.g. detoxification of pertussis toxin by chemical and/or genetic means (30)).
  • diphtheria antigen is included in the composition it is preferred also to include tetanus antigens and pertussis antigens. Similar, where a tetanus antigen is include it is preferred also to include diphtheria and pertussis antigens. Similar, where pertussis antigen is included it is preferred also to include diphtheria and tetanus antigens.
  • the pharmaceutical compositions containing the compounds described herein can include additives such as excipients.
  • Suitable pharmaceutically acceptable excipients include processing agents and drug delivery modifiers and enhancers, such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, hydroxypropyl- ⁇ -cyclodextrin, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of any two or more thereof.
  • processing agents and drug delivery modifiers and enhancers such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, hydroxypropyl- ⁇ -cyclodextrin, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of any two or
  • compositions containing the compounds of the invention may be in any form suitable for the intended method of administration, including, for example, a solution, a suspension, or an emulsion.
  • Liquid carriers are typically used in preparing solutions, suspensions, and emulsions.
  • Liquid carriers contemplated for use in the practice of the present invention include, for example, water, saline, pharmaceutically acceptable organic solvent(s), pharmaceutically acceptable oils or fats, and the like, as well as mixtures of two or more thereof.
  • the liquid carrier may contain other suitable pharmaceutically acceptable additives such as solubilizers, emulsifiers, nutrients, buffers, preservatives, suspending agents, thickening agents, viscosity regulators, stabilizers, and the like.
  • Suitable organic solvents include, for example, monohydric alcohols, such as ethanol, and polyhydric alcohols, such as glycols.
  • Suitable oils include, for example, soybean oil, coconut oil, olive oil, saffiower oil, cottonseed oil, and the like.
  • the carrier can also be an oily ester such as ethyl oleate, isopropyl myristate, and the like.
  • Compositions of the present invention may also be in the form of microparticles, microcapsules, and the like, as well as combinations of any two or more thereof. [00437]
  • the compounds and combinations of the present invention can also be administered in the form of liposomes.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are fonned by mono- or multilamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients, and the like.
  • the preferred lipids are the phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art.
  • compositions comprising an antigen, an antigenic CpG-ODN and a polycationic polymer.
  • Other immunostimulatory additives described in the art may be used, for example, as described in U.S. Patent No. 5,026,546; U.S. Patent No. 4,806,352; and U.S. Patent No. 5,026,543.
  • SMIP compounds as described in USSN 10/814480 and 60/582654 are contemplated as effective co ⁇ administration agents or combination with the compositions of the instant invention.
  • a controlled release delivery system may be used, such as a diffusion controlled matrix system or an erodible system, as described for example in: Lee, "Diffusion-Controlled Matrix Systems", pp.
  • the matrix may be, for example, a biodegradable material that can degrade spontaneously in situ and in vivo for, example, by hydrolysis or enzymatic cleavage, e.g., by proteases.
  • the delivery system may be, for example, a naturally occurring or synthetic polymer or copolymer, for example in the form of a hydrogel.
  • Exemplary polymers with cleavable linkages include polyesters, polyorthoesters, polyanhydrides, polysaccharides, poly(phosphoesters), polyamides, polyurethanes, poly(imidocarbonates) and poly(phosphazenes) .
  • the compounds of the invention may be administered enterally, orally, parenterally, sublingually, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired.
  • suitable modes of administration include oral, subcutaneous, transdermal, transmucosal, iontophoretic, intravenous, intramuscular, intraperitoneal, intranasal, subdermal, rectal, and the like.
  • Topical administration may also involve the use of transdermal administration such as transdermal patches or ionophoresis devices.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-propanediol.
  • a nontoxic parenterally acceptable diluent or solvent for example, as a solution in 1,3-propanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • a suitable nonirritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, cyclodextrins, and sweetening, flavoring, and perfuming agents.
  • Effective amounts of the compounds of the invention generally include any amount sufficient to detectably treat the disorders described herein.
  • Successful treatment of a subject in accordance with the invention may result in the inducement of a reduction or alleviation of symptoms in a subject afflicted with a medical or biological disorder to, for example, halt the further progression of the disorder, or the prevention of the disorder.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy.
  • compositions of the invention may be administered in conjunction with one or more antigens for use in therapeutic, prophylactic, or diagnostic methods of the present invention.
  • Preferred antigens include those listed below.
  • the compositions of the present invention may be used to treat or prevent infections caused by any of the below-listed microbes.
  • the compositions of the invention may also be combined with an adjuvant as described herein.
  • Antigens for use with the invention include, but are not limited to, one or more of the following antigens set forth below, or antigens derived from one or more of the pathogens set forth below:
  • Bacterial antigens suitable for use in the invention include proteins, polysaccharides, lipopolysaccharides, and outer membrane vesicles which may be isolated, purified or derived from a bacteria.
  • bacterial antigens may include bacterial lysates and inactivated bacteria formulations.
  • Bacteria antigens may be produced by recombinant expression.
  • Bacterial antigens preferably include epitopes which are exposed on the surface of the bacteria during at least one stage of its life cycle. Bacterial antigens are preferably conserved across multiple serotypes.
  • Bacterial antigens include antigens derived from one or more of the bacteria set forth below as well as the specific antigens examples identified below.
  • Meningitides antigens may include proteins (such as those identified in References 1 - 7), saccharides (including a polysaccharide, oligosaccharide or lipopolysaccharide), or outer-membrane vesicles (References 8, 9, 10, 11) purified or derived from N. meningitides serogroup A, C, W135, Y, and/or B. Meningitides protein antigens may be selected from adhesions, autotransporters, toxins, Fe acquisition proteins, and membrane associated proteins (preferably integral outer membrane protein).
  • Streptococcus pneumoniae antigens may include a saccharide (including a polysaccharide or an oligosaccharide) or protein from Streptococcus pneumoniae. Saccharide antigens may be selected from serotypes 1, 2, 3, 4, 5, 6B, 7F, 8, 9 ⁇ , 9V, 1OA, HA, 12F, 14, 15B, 17F, 18C, 19A, 19F, 20, 22F, 23F, and 33F. Protein antigens may be selected from a protein identified in WO 98/18931, WO 98/18930, US Patent No. 6,699,703, US Patent No. 6,800,744, WO 97/43303, and WO 97/37026.
  • Streptococcus pneumoniae proteins may be selected from the Poly Histidine Triad family (PhtX), the Choline Binding Protein family (CbpX), CbpX truncates, LytX family, LytX truncates, CbpX truncate-LytX truncate chimeric proteins, pneumolysin (Ply), PspA, PsaA, Spl28, SpIOl, Spl30, Spl25 or Spl33.
  • PhtX Poly Histidine Triad family
  • CbpX Choline Binding Protein family
  • CbpX truncates CbpX truncates
  • LytX family LytX truncates
  • pneumolysin (Ply) PspA, PsaA, Spl28, SpIOl, Spl30, Spl25 or Spl33.
  • Streptococcus pyogenes Group A Streptococcus antigens may include a protein identified in WO 02/34771 or WO 2005/032582 (including GAS 40), fusions of fragments of GAS M proteins (including those described in WO 02/094851, and Dale, Vaccine (1999) 17:193-200, and Dale, Vaccine 14(10): 944-948), fibronectin binding protein (Sfbl), Streptococcal heme- associated protein (Shp), and Streptolysin S (SagA).
  • Moraxella catarrhalis Moraxella antigens include antigens identified in WO 02/18595 and WO 99/58562, outer membrane protein antigens (HMW-OMP), C- antigen, and/or LPS.
  • Bordetella pertussis Pertussis antigens include petussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B. pertussis, optionally also combination with pertactin and/or agglutinogens 2 and 3 antigen.
  • Staphylococcus aureus Staph aureus antigens include S.
  • aureus type 5 and 8 capsular polysaccharides optionally conjugated to nontoxic recombinant Pseudomonas aeruginosa exotoxin A, such as StaphVAXTM, or antigens derived from surface proteins, invasins (leukocidin, kinases, hyaluronidase), surface factors that inhibit phagocytic engulfment (capsule, Protein A), carotenoids, catalase production, Protein A, coagulase, clotting factor, and/or membrane-damaging toxins (optionally detoxified) that lyse eukaryotic cell membranes (hemolysins, leukotoxin, leukocidin).
  • Pseudomonas aeruginosa exotoxin A such as StaphVAXTM
  • antigens derived from surface proteins invasins (leukocidin, kinases, hyaluronidase), surface factors that inhibit
  • Staphylococcus epidermis S. epidermidis antigens include slime-associated antigen (SAA). Tetanus: Tetanus antigens include tetanus toxoid (TT), preferably used as a carrier protein in conjunction/conjugated with the compositions of the present invention.
  • Tetanus antigens include diphtheria toxin, preferably detoxified, such as CRMi 97 , additionally antigens capable of modulating, inhibiting or associated with ADP ribosylation are contemplated for combination/co- adrninistration/conjugation with the compositions of the present invention, the diphtheria toxoids are preferably used as carrier proteins.
  • Haemophilus influenzae B Hib antigens include a Hib saccharide antigen.
  • Pseudomonas aeruginosa Pseudomonas antigens include endotoxin A, Wzz protein, P. aeruginosa LPS, more particularly LPS isolated from PAOl (05 serotype), and/or Outer Membrane Proteins, including Outer Membrane Proteins F (OprF) (Infect Imm ⁇ n. 2001 May; 69(5): 3510-3515).
  • Legionella pneumophila (Legionnairs' Disease): L. pneumophila antigens may optionally derived from cell lines with disrupted asd genes (Infect Immun.
  • Streptococcus agalactiae Group B Streptococcus: Group B Streptococcus antigens include a protein or saccharide antigen identified in WO 02/34771, WO 03/093306, WO 04/041157, or WO 2005/002619 (including proteins GBS 80, GBS 104, GBS 276 and GBS 322, and including saccharide antigens derived from serotypes Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII).
  • Neiserria gonorrhoeae antigens include Por (or porin) protein, such as PorB (see Zhu et al, Vaccine (2004) 22:660 - 669), a transferring binding protein, such as TbpA and TbpB (See Price et al, Infection and Immunity (2004) 71(1):277 - 283), a opacity protein (such as Opa), a reduction-modifiable protein (Rmp), and outer membrane vesicle (OMV) preparations (see Plante et al, J Infectious Disease (2000) 182:848 - 855), also see e.g.
  • PorB PorB
  • TbpA and TbpB See Price et al, Infection and Immunity (2004) 71(1):277 - 283
  • a opacity protein such as Opa
  • Rmp reduction-modifiable protein
  • OMV outer membrane vesicle
  • Chlamydia trachomatis antigens include antigens derived from serotypes A, B, Ba and C are (agents of trachoma, a cause of blindness), serotypes L 1 , L 2 & L 3 (associated with Lymphogranuloma venereum), and serotypes, D-K. Chlamydia trachomas antigens may also include an antigen identified in WO 00/37494, WO 03/049762, WO 03/068811, or WO 05/002619.
  • Treponema pallidum Syphilis antigens include TmpA antigen.
  • Haemophilus ducreyi causing chancroid
  • Ducreyi antigens include outer membrane protein (DsrA).
  • Enterococcus faecalis or Enter ococcus faecium Antigens include a trisaccharide repeat or other Enterococcus derived antigens provided in US Patent No. 6,756,361.
  • Helicobacter pylori H pylori antigens include Cag, Vac, Nap, HopX, Hop Y and/or urease antigen.
  • Staphylococcus saprophyticus Antigens include the 160 kDa hemagglutinin of S. saprophyticus antigen.
  • Yersinia enterocolitica Antigens include LPS (Infect Immun. 2002 August; 70(8): 4414).
  • E. coli E. coli antigens may be derived from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), and/or enterohemorrhagic E. coli (EHEC).
  • Bacillus anthracis anthrax: B.
  • anthracis antigens are optionally detoxified and may be selected from A-components (lethal factor (LF) and edema factor (EF)), both of which can share a common B-component known as protective antigen (PA).
  • LF lethal factor
  • EF edema factor
  • PA protective antigen
  • Plague antigens include Fl capsular antigen (Infect Immun. 2003 Jan; 71(1)): 374-383, LPS (Infect Immun. 1999 Oct; 67(10): 5395), Yersinia pestis V antigen (Infect Immun. 1997 Nov; 65(11): 4476-4482).
  • Tuberculosis antigens include lipoproteins, LPS, BCG antigens, a fusion protein of antigen 85B (Ag85B) and/or ESAT-6 optionally formulated in cationic lipid vesicles (Infect Immun. 2004 October; 72(10): 6148), Mycobacterium tuberculosis (Mtb) isocitrate dehydrogenase associated antigens (Proc Natl Acad Sd USA. 2004 Aug 24; 101(34): 12652), and/or MPT51 antigens ⁇ Infect Immun. 2004 July; 72(7): 3829).
  • Antigens include outer membrane proteins, including the outer membrane protein A and/or B (OmpB) ⁇ Biochim Biophys Acta. 2004 Nov l;1702(2):145), LPS, and surface protein antigen (SPA) (J Autoimmun. 1989 Jun;2 Suppl:81).
  • Listeria monocytogenes Antigens derived from L. monocytogenes are preferably used as carriers/vectors for intracytoplasmic delivery of conjugates/associated compositions of the present invention.
  • Chlamydia pneumoniae Antigens include those identified in WO 02/02606.
  • Vibrio cholerae Antigens include proteinase antigens, LPS, particularly lipopolysaccharides of Vibrio cholerae II, Ol Inaba O-specif ⁇ c polysaccharides, V. cholera 0139, antigens of IEM108 vaccine (Infect Immun. 2003 Oct;71(10):5498- 504), and/or Zonula occludens toxin (Zot).
  • Salmonella typhi typhoid fever
  • Antigens include capsular polysaccharides preferably conjugates (Vi, i.e. vax-TyVi).
  • Antigens include lipoproteins (such as OspA, OspB, Osp C and Osp D), other surface proteins such as OspE-related proteins (Erps), decorin-binding proteins (such as DbpA), and antigenically variable VI proteins. , such as antigens associated with P39 and P13 (an integral membrane protein, Infect Immun.2001 May; 69(5): 3323-3334), VlsE Antigenic Variation Protein (J Clin Microbiol. 1999 Dec; 37(12): 3997).
  • Porphyromonas gingivalis Antigens include P. gingivalis outer membrane protein (OMP).
  • Antigens include an OMP, including OMP A, or a polysaccharide optionally conjugated to tetanus toxoid.
  • further bacterial antigens of the invention may be capsular antigens, polysaccharide antigens or protein antigens of any of the above.
  • Further bacterial antigens may also include an outer membrane vesicle (OMV) preparation.
  • OMV outer membrane vesicle
  • antigens include live, attenuated, split, and/or purified versions of any of the aforementioned bacteria.
  • the bacterial or microbial derived antigens of the present invention may be gram-negative or gram-positive and aerobic or anaerobic.
  • any of the above bacterial-derived saccharides can be conjugated to another agent or antigen, such as a carrier protein (for example CRM 197 ).
  • a carrier protein for example CRM 197
  • Such conjugation may be direct conjugation effected by reductive animation of carbonyl moieties on the saccharide to amino groups on the protein, as provided in US Patent No. 5,360,897 and Can JBiochem Cell Biol. 1984 May; 62(5): 270-5.
  • the saccharides can be conjugated through a linker, such as, with succinamide or other linkages provided in Bioconjugate Techniques, 1996 and CRC, Chemistry of Protein Conjugation and Cross-Linking, 1993.
  • Viral Antigens suitable for use in the invention include inactivated (or killed) virus, attenuated virus, split virus formulations, purified subunit formulations, viral proteins which may be isolated, purified or derived from a virus, and Virus Like Particles (VLPs).
  • Viral antigens may be derived from viruses propagated on cell culture or expressed recombinantly.
  • Viral antigens preferably include epitopes which are exposed on the surface of the virus during at least one stage of its life cycle. Viral antigens are preferably conserved across multiple serotypes.
  • Viral antigens include antigens derived from one or more of the viruses set forth below as well as the specific antigens examples identified below.
  • Orthomyxovirus Viral antigens may be derived from an Orthomyxovirus, such as Influenza A, B and C.
  • Orthomyxovirus antigens may be selected from one or more of the viral proteins, including hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), matrix protein (Ml), membrane protein (M2), one or more of the transcriptase components (PBl, PB2 and PA).
  • Preferred antigens include HA and NA.
  • Influenza antigens may be derived from interpandemic (annual) flu strains.
  • influenza antigens may be derived from strains with the potential to cause pandemic a pandemic outbreak (i.e., influenza strains with new haemagglutinin compared to the haemagglutinin in currently circulating strains, or influenza strains which are pathogenic in avian subjects and have the potential to be transmitted horizontally in the human population, or influenza strains which are pathogenic to humans).
  • Paramyxoviridae viruses Viral antigens may be derived from Paramyxoviridae viruses, such as Pneumoviruses (RSV), Paramyxoviruses (PIV) and Morbilliviruses (Measles).
  • Viral antigens may be derived from a Pneumovirus, such as Respiratory syncytial virus (RSV), Bovine respiratory syncytial virus, Pneumonia virus of mice, and Turkey rhinotracheitis virus.
  • the Pneumovirus is RSV.
  • Pneumovirus antigens may be selected from one or more of the following proteins, including surface proteins Fusion (F), Glycoprotein (G) and Small Hydrophobic protein (SH), matrix proteins M and M2, nucleocapsid proteins N, P and L and nonstructural proteins NSl and NS2.
  • Preferred Pneumovirus antigens include F, G and M. See e.g., J Gen Virol.
  • Pneumovirus antigens may also be formulated in or derived from chimeric viruses.
  • chimeric RSV/PIV viruses may comprise components of both RSV and PIV.
  • Paramyxovirus Viral antigens may be derived from a Paramyxovirus, such as Parainfluenza virus types 1 - 4 (PIV), Mumps, Sendai viruses, Simian virus 5, Bovine parainfluenza virus and Newcastle disease virus.
  • the Paramyxovirus is PIV or Mumps.
  • Paramyxovirus antigens may be selected from one or more of the following proteins: Hemagglutinin -Neuraminidase (HN), Fusion proteins Fl and F2, Nucleoprotein (NP), Phosphoprotein (P), Large protein (L), and Matrix protein (M).
  • HN Hemagglutinin -Neuraminidase
  • NP Nucleoprotein
  • Phosphoprotein P
  • Large protein L
  • M Matrix protein
  • Preferred Paramyxovirus proteins include HN, Fl and F2.
  • Paramyxovirus antigens may also be formulated in or derived from chimeric viruses.
  • chimeric RSV/PIV viruses may comprise components of both RSV and PIV.
  • Commercially available mumps vaccines include live attenuated mumps virus, in either a monovalent form or in combination with measles and rubella vaccines (MMR).
  • Morbillivirus Viral antigens may be derived from a Morbillivirus, such as Measles. Morbillivirus antigens may be selected from one or more of the following proteins: hemagglutinin (H), Glycoprotein (G), Fusion factor (F), Large protein (L), Nucleoprotein (NP), Polymerase phosphoprotein (P), and Matrix (M). Commercially available measles vaccines include live attenuated measles virus, typically in combination with mumps and rubella (MMR).
  • Picornavirus Viral antigens may be derived from Picornaviruses, such as Enteroviruses, Rhinoviruses, Heparnavirus, Cardioviruses and Aphthoviruses.
  • Antigens derived from Enteroviruses are preferred.
  • Enterovirus Viral antigens may be derived from an Enterovirus, such as Poliovirus types 1, 2 or 3, Coxsackie A virus types 1 to 22 and 24, Coxsackie B virus types 1 to 6, Echovirus (ECHO) virus) types 1 to 9, 11 to 27 and 29 to 34 and Enterovirus 68 to 71.
  • the Enterovirus is poliovirus.
  • Enterovirus antigens are preferably selected from one or more of the following Capsid proteins VPl, VP2, VP3 and VP4.
  • Commercially available polio vaccines include Inactivated Polio Vaccine (IPV) and Oral poliovirus vaccine (OPV).
  • Heparnavirus Viral antigens may be derived from an Heparnavirus, such as Hepatitis A virus (HAV). Commercially available HAV vaccines include inactivated HAV vaccine.
  • Togavirus Viral antigens may be derived from a Togavirus, such as a Rubivirus, an Alphavirus, or an Arterivirus. Antigens derived from Rubivirus, such as Rubella virus, are preferred. Togavirus antigens may be selected from El, E2, E3, C, NSP-I, NSPO-2, NSP-3 or NSP-4. Togavirus antigens are preferably selected from El, E2 or E3.
  • Rubella vaccines include a live cold- adapted virus, typically in combination with mumps and measles vaccines (MMR).
  • Flavivirus Viral antigens may be derived from a Flavivirus, such as Tick- borne encephalitis (TBE), Dengue (types 1, 2, 3 or 4), Yellow Fever, Japanese encephalitis, West Nile encephalitis, St. Louis encephalitis, Russian spring-summer encephalitis, Powassan encephalitis.
  • Flavivirus antigens may be selected from PrM, M, C, E, NS-I, NS-2a, NS2b, NS3, NS4a, NS4b, and NS5.
  • Flavivirus antigens are preferably selected from PrM, M and E.
  • Commercially available TBE vaccine include inactivated virus vaccines.
  • Pestivirus Viral antigens may be derived from a Pestivirus, such as Bovine viral diarrhea (BVDV), Classical swine fever (CSFV) or Border disease (BDV).
  • Hepadnavirus Viral antigens may be derived from a Hepadnavirus, such as Hepatitis B virus. Hepadnavirus antigens may be selected from surface antigens (L, M and S), core antigens (HBc, HBe).
  • Commercially available HBV vaccines include subunit vaccines comprising the surface antigen S protein.
  • Hepatitis C virus Viral antigens may be derived from a Hepatitis C virus (HCV). HCV antigens may be selected from one or more of El, E2, E1/E2, NS345 polyprotein, NS 345-core polyprotein, core, and/or peptides from the nonstructural regions (Houghton et al., Hepatology (1991) 14:381).
  • Rhabdovirus Viral antigens may be derived from a Rhabdovirus, such as a Lyssavirus (Rabies virus) and Vesiculovirus (VSV). Rhabdovirus antigens may be selected from glycoprotein (G), nucleoprotein (N), large protein (L), nonstructural proteins (NS).
  • Rabies virus vaccine comprise killed virus grown on human diploid cells or fetal rhesus lung cells.
  • Caliciviridae Viral antigens may be derived from Calciviridae, such as Norwalk virus.
  • Coronavirus Viral antigens may be derived from a Coronavirus, SARS, Human respiratory coronavirus, Avian infectious bronchitis (IBV), Mouse hepatitis virus (MHV), and Porcine transmissible gastroenteritis virus (TGEV).
  • Coronavirus antigens may be selected from spike (S), envelope (E), matrix (M), nucleocapsid (N), and Hemagglutinin-esterase glycoprotein (HE).
  • the Coronavirus antigen is derived from a SARS virus.
  • Retrovirus Viral antigens may be derived from a Retrovirus, such as an Oncovirus, a Lentivirus or a Spumavirus. Oncovirus antigens may be derived from HTLV-I, HTLV-2 or HTLV-5. Lentivirus antigens may be derived from HIV-I or HIV-2. Retrovirus antigens may be selected from gag, pol, env, tax, tat, rex, rev, nef, vif, vpu, and vpr.
  • HIV antigens may be selected from gag (p24gag and p55gag), env (gpl60 and gp41), pol, tat, nef, rev vpu, miniproteins, (preferably p55 gag and gpl40v delete). HIV antigens may be derived from one or more of the following strains: fflV ⁇ ib, HIVsF 2 , HIVLAV, HIV LA I, HIVMN, HIV-1 C M235, HIV-1 US4 .
  • Reovirus Viral antigens may be derived from a Reovirus, such as an Orthoreo virus, a Rotavirus, an Orbivirus, or a Coltivirus.
  • Reovirus antigens may be selected from structural proteins ⁇ l, ⁇ 2, ⁇ 3, ⁇ l, ⁇ 2, ⁇ l, ⁇ 2, or ⁇ 3, or nonstructural proteins ⁇ NS, ⁇ NS, or ⁇ ls.
  • Preferred Reovirus antigens may be derived from a Rotavirus.
  • Rotavirus antigens may be selected from VPl, VP2, VP3, VP4 (or the cleaved product VP5 and VP8), NSP 1, VP6, NSP3, NSP2, VP7, NSP4, or NSP5.
  • Preferred Rotavirus antigens include VP4 (or the cleaved product VP5 and VP8), and VP7.
  • Parvovirus Viral antigens may be derived from a Parvovirus, such as Parvovirus B19. Parvovirus antigens may be selected from VP-I, VP-2, VP-3, NS-I and NS-2. Preferably, the Parvovirus antigen is capsid protein VP-2. Delta hepatitis virus (HDV): Viral antigens may be derived HDV, particularly ⁇ -antigen from HDV (see, e.g., U.S. Patent No. 5,378,814). Hepatitis E virus (HEV): Viral antigens may be derived from HEV. Hepatitis G virus (HGV): Viral antigens may be derived from HGV.
  • HDV Delta hepatitis virus
  • HEV Hepatitis E virus
  • HEV Hepatitis G virus
  • Human Herpesvirus Viral antigens may be derived from a Human Herpesvirus, such as Herpes Simplex Viruses (HSV), Varicella-zoster virus (VZV), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Human Herpesvirus 6 (HHV6), Human Herpesvirus 7 (HHV7), and Human Herpesvirus 8 (HHV8).
  • Human Herpesvirus antigens may be selected from immediate early proteins ( ⁇ ), early proteins ( ⁇ ), and late proteins ( ⁇ ).
  • HSV antigens may be derived from HSV-I or HSV-2 strains.
  • HSV antigens may be selected from glycoproteins gB, gC, gD and gH, fusion protein (gB), or immune escape proteins (gC, gE, or gl).
  • VZV antigens may be selected from core, nucleocapsid, tegument, or envelope proteins.
  • a live attenuated VZV vaccine is commercially available.
  • EBV antigens may be selected from early antigen (EA) proteins, viral capsid antigen (VCA), and glycoproteins of the membrane antigen (MA).
  • CMV antigens may be selected from capsid proteins, envelope glycoproteins (such as gB and gH), and tegument proteins Papovaviruses: Antigens may be derived from Papovaviruses, such as Papillomaviruses and Polyomaviruses. Papillomaviruses include HPV serotypes 1, 2, 4, 5, 6, 8, 11, 13, 16, 18, 31, 33, 35, 39, 41, 42, 47, 51, 57, 58, 63 and 65. Preferably, HPV antigens are derived from serotypes 6, 11, 16 or 18. HPV antigens maybe selected from capsid proteins (Ll) and (LT), or El - E7, or fusions thereof.
  • HPV antigens are preferably formulated into virus-like particles (VLPs).
  • Polyomyavirus viruses include BK virus and JK virus.
  • Polyomavirus antigens may be selected from VPl, VP2 or VP3. Further provided are antigens, compositions, methods, and microbes included in Vaccines, 4 th Edition (Plotkin and Orenstein ed. 2004); Medical Microbiology 4 th Edition (Murray et al. ed. 2002); Virology, 3rd Edition (W.K. Joklik ed. 1988); Fundamental Virology, 2nd Edition (B.N. Fields and D.M. Knipe, eds. 1991), which are contemplated in conjunction with the compositions of the present invention.
  • compositions of the present invention are combined with fungal antigens for use in methods of the present invention, including treatment or prevention of mycoses.
  • Fungal antigens for use herein, associated with vaccines include those described in: U.S. Pat. Nos. 4,229,434 and 4,368,191 for prophylaxis and treatment of trichopytosis caused by Trichophyton mentagrophytes; U.S. Pat. Nos. 5,277,904 and 5,284,652 for a broad spectrum dermatophyte vaccine for the prophylaxis of dermatophyte infection in animals, such as guinea pigs, cats, rabbits, horses and lambs, these antigens comprises a suspension of killed T.
  • fungal antigens for use herein may be derived from Dermatophytres, including: Epidermophytonfloccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T.
  • Fungal pathogens for use as antigens or in derivation of antigens in conjunction with the compositions of the present invention comprise Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, Aspergillus terreus, Aspergillus sydowi, Aspergillus flavatus, Aspergillus glaucus, Blastoschizomyces capitatus, Candida albicans, Candida enolase, Candida tropicalis, Candida glabrata, Candida krusei, Candida par aps ⁇ losis, Candida stellatoidea, Candida kusei, Candida parakwsei, Candida lusitaniae, Candida pseudotropicalis, Candida guilliermondi, Cladospor
  • fungi from which antigens are derived include Alternaria spp, Curvularia spp, Helminthosporium spp, Fusarium spp, Aspergillus spp, Penicillium spp, Monolinia spp, Rhizoctonia spp, Paecilomyces spp, Pithomyces spp, and Cladosporium spp. Processes for producing a fungal antigens are well known in the art (see US Patent No. 6,333,164).
  • a solubilized fraction extracted and separated from an insoluble fraction obtainable from fungal cells of which cell wall has been substantially removed or at least partially removed characterized in that the process comprises the steps of: obtaining living fungal cells; obtaining fungal cells of which cell wall has been substantially removed or at least partially removed; bursting the fungal cells of which cell wall has been substantially removed or at least partially removed; obtaining an insoluble fraction; and extracting and separating a solubilized fraction from the insoluble fraction.
  • Embodiments of the invention include compositions and methods related to a prophylactic and therapeutic treatments for microbes that can be neutralized prior to infection of a cell.
  • microbes bacteria, viruses and/or fungi
  • STDs sexually transmitted diseases
  • compositions are combined with antigens derived from a viral or bacterial STD.
  • Antigens derived from bacteria or viruses can be administered in conjunction with the compositions of the present invention to provide protection against at least one of the following STDs, among others: chlamydia, genital herpes, hepatitis (particularly HCV), genital warts, gonorrhea, syphilis and/or chancroid (See, WO00/15255).
  • compositions of the present invention are co ⁇ administered with an antigen for the prevention or treatment of an STD.
  • Antigens derived from the following viruses associated with STDs, which are described in greater detail above, are preferred for co-administration with the compositions of the present invention: hepatitis (particularly HCV), HPV, HIV, or HSV.
  • antigens derived from the following bacteria associated with STDs which are described in greater detail above, are preferred for co-administration with the compositions of the present invention: Neiserria gonorrhoeae, Chlamydia pneumoniae, Chlamydia trachomatis, Treponema pallidum, ox Haemophilus ducreyi.
  • the invention provides methods of preventing and/or treating infection by a respiratory pathogen, including a virus, bacteria, or fungi such as respiratoiy syncytial virus (RSV), PIV, SARS virus, influenza, Bacillus anthracis, particularly by reducing or preventing infection and/or one or more symptoms of respiratory virus infection.
  • a respiratory pathogen including a virus, bacteria, or fungi such as respiratoiy syncytial virus (RSV), PIV, SARS virus, influenza, Bacillus anthracis
  • a composition comprising an antigen described herein, such as one derived from a respiratory virus, bacteria or fungus is administered in conjunction with the compositions of the present invention to an individual which is at risk of being exposed to that particular respiratory microbe, has been exposed to a respiratory microbe or is infected with a respiratory virus, bacteria or fungus.
  • the composition(s) of the present invention is/are preferably co-administered at the same time or in the same formulation with an anti
  • Tumor Antigens can be, for example, peptide-containing tumor antigens, such as a polypeptide tumor antigen or glycoprotein tumor antigens.
  • a tumor antigen can also be, for example, a saccharide-containing tumor antigen, such as a glycolipid tumor antigen or a ganglioside tumor antigen.
  • the tumor antigen can further be, for example, a polynucleotide-containing tumor antigen that expresses a polypeptide-containing tumor antigen, for instance, an RNA vector construct or a DNA vector construct, such as plasmid DNA.
  • Tumor antigens appropriate for the practice of the present invention encompass a wide variety of molecules, such as (a) polypeptide-containing tumor antigens, including polypeptides (which can range, for example, from 8-20 amino acids in length, although lengths outside this range are also common), lipopolypeptides and glycoproteins, (b) saccharide-containing tumor antigens, including poly-saccharides, mucins, gangliosides, glycolipids and glycoproteins, and (c) polynucleotides that express antigenic polypeptides.
  • the tumor antigens can be, for example, (a) full length molecules associated with cancer cells, (b) homologs and modified forms of the same, including molecules with deleted, added and/or substituted portions, and (c) fragments of the same.
  • Tumor antigens can be provided in recombinant form.
  • Tumor antigens include, for example, class I-restricted antigens recognized by CD8+ lymphocytes or class Il-restricted antigens recognized by CD4+ lymphocytes.
  • tumor antigens are known in the art, including: (a) cancer-testis antigens such as NY-ESO-I, SSX2, SCPl as well as RAGE, BAGE, GAGE and MAGE family polypeptides, for example, GAGE-I, GAGE-2, MAGE-I, MAGE-2, MAGE-3, MAGE-4, MAGE-5, MAGE-6, and MAGE-12 (which can be used, for example, to address melanoma, lung, head and neck, NSCLC, breast, gastrointestinal, and bladder tumors), (b) mutated antigens, for example, p53 (associated with various solid tumors, e.g., colorectal, lung, head and neck cancer), p21/Ras (associated with, e.g., melanoma, pancreatic cancer and colorectal cancer), CDK4 (associated with, e.g., melanoma), MUMl (associated with, e.g., melanoma), caspase
  • Additional tumor antigens which are known in the art include pi 5, Hom/Mel- 40, H-Ras, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens, including E6 and E7, hepatitis B and C virus antigens, human T-cell lymphotropic virus antigens, TSP-180, pl85erbB2, pl80erbB-3, c-met, mn-23Hl, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K- ras, p 16, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1, CO-029, F
  • Polynucleotide-containing antigens in accordance with the present invention typically comprise polynucleotides that encode polypeptide cancer antigens such as those listed above.
  • Preferred polynucleotide-containing antigens include DNA or RNA vector constructs, such as plasmid vectors (e.g., pCMV), which are capable of expressing polypeptide cancer antigens in vivo.
  • Tumor antigens may be derived, for example, from mutated or altered cellular components. After alteration, the cellular components no longer perfo ⁇ n their regulatory functions, and hence the cell may experience uncontrolled growth.
  • altered cellular components include ras, p53, Rb, altered protein encoded by the Wilms' tumor gene, ubiquitin, mucin, protein encoded by the DCC, APC, and MCC genes, as well as receptors or receptor-like structures such as neu, thyroid hormone receptor, platelet derived growth factor (PDGF) receptor, insulin receptor, epidermal growth factor (EGF) receptor, and the colony stimulating factor (CSF) receptor.
  • PDGF platelet derived growth factor
  • EGF epidermal growth factor
  • CSF colony stimulating factor
  • carrier proteins such as CRMi 9 - 7 , tetanus toxoid, or Salmonella typhimurium antigen can be used in conjunction/conjugation with compounds of the present invention for treatment of cancer.
  • the cancer antigen combination therapies will show increased efficacy and bioavailability as compared with existing therapies. Additional information on cancer or tumor antigens can be found, for example, in Moingeon P, "Cancer vaccines," Vaccine, 2001, 19:1305-1326; Rosenberg SA, "Progress in human tumor immunology and immunotherapy,” Nature, 2001, 411:380- 384; Dermine, S.
  • compositions of the present invention are used in conjunction with an antigen for treatment of a pediatric population, as in a pediatric antigen.
  • the pediatric population is less than about 3 years old, or less than about 2 years, or less than about 1 years old.
  • the pediatric antigen (in conjunction with the composition of the present invention) is administered multiple times over at least 1, 2, or 3 years.
  • the compositions of the present invention are used in conjunction with an antigen for treatment of a geriatric population, as in a geriatric antigen.
  • antigens for use in conjunction with the compositions of the present include hospital acquired (nosocomial) associated antigens.
  • parasitic antigens are contemplated in conjunction with the compositions of the present invention. Examples of parasitic antigens include those derived from organisms causing malaria and/or Lyme disease.
  • the antigens in conjunction with the compositions of the present invention are associated with or effective against a mosquito born illness.
  • the antigens in conjunction with the compositions of the present invention are associated with or effective against encephalitis.
  • the antigens in conjunction with the compositions of the present invention are associated with or effective against an infection of the nervous system.
  • the antigens in conjunction with the compositions of the present invention are antigens transmissible through blood or body fluids.
  • the methods comprise: (a) providing an emulsion by dispersing a mixture comprising (i) water, (ii) a detergent, (iii) an organic solvent, and (iv) a biodegradable polymer selected from the group consisting of a poly( ⁇ -hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate.
  • the polymer is typically present in the mixture at a concentration of about 1% to about 30% relative to the organic solvent, while the detergent is typically present in the mixture at a weight-to- weight detergent-to-polymer ratio of from about 0.00001:1 to about 0.1:l (more typically about 0.0001:1 to about 0.1:1, about 0.001:1 to about 0.1:1, or about 0.005:1 to about 0.1:1); (b) removing the organic solvent from the emulsion; and (c) adsorbing an antigen on the surface of the microparticles.
  • the biodegradable polymer is present at a concentration of about 3% to about 10% relative to the organic solvent.
  • Microparticles for use herein will be formed from materials that are sterilizable, non-toxic and biodegradable. Such materials include, without limitation, poly( ⁇ -hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate.
  • microparticles for use with the present invention are derived from a poly( ⁇ ;-hydroxy acid), in particular, from a poly(lactide) ("PLA”) or a copolymer of D,L-lactide and glycolide or glycolic acid, such as a poly(D,L-lactide-co-glycolide) (“PLG” or "PLGA”), or a copolymer of D,L-lactide and caprolactone.
  • PLA poly(lactide)
  • PLA poly(lactide)
  • PLA poly(D,L-lactide-co-glycolide)
  • caprolactone a copolymer of D,L-lactide and caprolactone
  • microparticles may be derived from any of various polymeric starting materials which have a variety of molecular weights and, in the case of the copolymers such as PLG, a variety of lactide:glycolide ratios, the selection of which will be largely a matter of choice, depending in part on the coadministered macromolecule. These parameters are discussed more fully below.
  • Further antigens may also include an outer membrane vesicle (OMV)
  • Antigen References The following references include antigens useful in conjunction with the
  • compositions and methods of the present invention are compositions and methods of the present invention.
  • halo is preferably chloro, bromo, or iodo.
  • the reaction is a Grignard, carried out in an inert organic solvent, such as toluene, at a temperature between room temperature and the reflux temperature of the reaction mixture. Most significantly, the reaction is dependent on solvent conditions.
  • the reaction When carried out in a Toluene:THF:ether solvent system, the reaction provides the product in high yield. The product is precipitated from the reaction mixture with ammonium chloride (NH 4 Cl).
  • NH 4 Cl ammonium chloride
  • the resulting bis-3,4(3'-indolyl)-lN-pyrrole-2,5-dione product may be isolated by standard techniques.
  • L is a good leaving group such as chloro, bromo, iodo, mesyl, tosyl, and the like.
  • L may also be a hydroxy or other precursor that may be readily converted to a good leaving group by techniques known in the art.
  • the hydroxy may be readily converted to a sulfonic ester such as mesyl by reacting the hydroxy with methanesulfonyl chloride to produce the mesylate leaving group.
  • the reaction is accomplished by any of the known methods of preparing N- substituted indoles.
  • This reaction usually involves approximately equimolar amounts of the two reagents, although other ratios, especially those wherein the alkylating reagent is in excess, are operative.
  • the reaction is best carried out in a polar aprotic solvent employing an alkali metal salt or other such alkylation conditions as are appreciated in the art.
  • a catalytic amount of iodide salt such as potassium iodide may be added to speed the reaction.
  • Reaction conditions include the following: Potassium hexamethyldisilazide in dimethylformamide or tetrahydrofuran, sodium hydride in dimethylformamide.
  • the reaction is carried out under slow reverse addition with cesium carbonate in either acetonitrile, dimethylformamide (DMF), or tetrahydrofuran (THF).
  • the temperature of the reaction is preferably from about ambient temperature to about the reflux temperature of the reaction mixture.
  • Scheme 1 is performed as a one pot procedure, with reagents in step a being NH 4 OH, CuCl in H 2 O.
  • reagents in step a being NH 4 OH, CuCl in H 2 O.
  • R 2 -R 5 are as defined herein.
  • R 9 as shown in scheme 3 is H, -OH, -CN, alkyl, aryl, heterocyclyl, alkoxy, or -NR a R b as defined herein. It is contemplated that the above structure may replace Formula I to allow substitution at Rg, whereby all other substituents are as defined herein.
  • Example 60 [00460] 1 -(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine dihydrochloride [00461] A mixture of 15.22 g (59.52 mmol) l-chloro-4-(4- pyridylmethyl)phthalazine (for preparation see German Auslegeschriftno. 1 061 788 published JuI. 23, 1959]), 7.73 g (60.59 mmol) 4-chloroaniline and 200 ml 1-butanol is heated for 2 h under reflux. The crystallizate which is obtained when the mixture slowly cools to 5° C. is then filtered off and washed with 1-butanol and ether.
  • the filter residue is dissolved in about 200 ml hot methanol, the solution is treated with 0.75 g activated carbon and filtered Via a Hyflo Super CeI, and the pH of the filtrate is adjusted to about 2.5 with 7 ml 3N methanolic HCl.
  • the filtrate is evaporated to about half the original volume and ether added until slight turbidity occurs; cooling then leads to the precipitation of crystals.
  • the crystallizate is filtered off, washed with a mixture of methanol/ether (1 :2) as well as ether, dried for 8 h at 110° C. under HV, and equilibrated for 72 h at 20° C. and in room atmosphere.
  • Example 62 1 -(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine hydrochloride [00465] A mixture of 1.28 g (5 mmol) l-chloro-4-(4-pyridylmethyl)phthalazine, 0.67 g (5.25 mmol) 4-chloroaniline and 15 ml 1-butanol is heated for 0.5 h at 100 C while stirring in a nitrogen atmosphere. The mixture is then cooled to RT, filtered, and the filtrate washed with 1-butanol and ether.
  • the crystallizate is dissolved in 40 ml of hot methanol, the solution treated with activated carbon, filtered via Hyflo Super CeI, and the filtrate evaporated to about half its original volume, resulting in the formation of a crystalline precipitate. After cooling to 0° C, filtration, washing of the filter residue with ether, and drying under HV for 8 h at 130° C, the title compound is obtained; m.p.
  • Example 63 1 -(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine
  • a mixture of 14.19 g (0.1 mol) phosphorus pentoxide, 13.77 g (0.1 mol) triethylamine hydrochloride and 12.76 g (0.1 mol) 4-chloroaniline is heated and stirred in a nitrogen atmosphere at 200° C. until a homogeneous melt has formed (about 20 min).
  • 5.93 g (0.025 mol) 4-(4-pyridylmethyl)-l (2H)- phthalazinone for preparation see German Auslegeschrift no.
  • 381 mg (0.77 mmol) 7-trifluoroacetamino-l- chloro- 4-(4-pyridylmethyl)phthalazine is heated to 100° C for 5 h in 3.1 ml n-butanol with 295 mg (2.31 mmol) 4-chloroaniline.
  • Example 65 5-(4-Chloroanilino)-8-(4-pyridylmethyl)pyrido[2.3-d]pyridazine [00476] Under N 2 atmosphere, a mixture of 1.19 g (8.38 mmol) phosphorus pentoxide, 1.156 g (8.4 mmol) triethylamine hydrochloride, and 1.072 g (8. 4 mmol) 4-chloroaniline is heated for 5 min to 200° C.
  • the starting material is prepared as follows: [00477] 65.1) 6-(Pyridin-4-yl)-l pyridin-5,7-dione [00478] To a suspension of 20.27 g (150 mmol) furo[3,4-b]pyridin-5(7h)-one (for preparation see Synthesis 1997,113) and 14.13 ml (150 mmol) 4- pyridinecarbaldehyde in 120 ml methanol and 75 ml ethyl propionate, 27.8 ml (150 mmol) of a 5.4M solution of sodium methylate in methanol is added dropwise under ice cooling (and N 2 atmosphere).
  • Example 67 A: l-(3-Phenoxyanilino)-4-(4-pyridylmethyl)phthalazine [00490] A mixture of 256 mg (1.00 mmol) l-chloro-4-(4- pyridylmethyl)phthalazine and 556 mg (3.00 mmol) 4-phenoxyaniline (Aldrich) is heated for 2 h at 90° C. The melt is cooled and stirred with 6 ml NH 3 solution (10% in water: or 10 ml sat. NaHCO 3 solution) and 15 ml dichloromethane/methanol 50:1 for 30 min. The aqueous phase is then separated off and extracted again with dichloromethane.
  • 200 mg (0.78 mmol) l-chloro-4-(4- pyridylmethyl)phthalazine (Example 67A.1), 173 mg (1.25 mmol) K 2 CO 3 , and 120 mg (0.94 mmol) 3-chlorophenol (Fluka) are heated in 2 ml DMSO for 3 h to 90° C.
  • the reaction mixture is distributed between 20 ml water and 20 ml ethyl acetate, and the aqueous phase separated and extracted with 2 portions of ethyl acetate.
  • HR is a nucleophile, such as ammonia and the base is triethylamine or NaH.
  • a phosphoramidite method with o-xylylene N,N-diethylphosphoramidite (XEPA) u is effective in this system (Scheme 4).
  • the isopropylidene and Boc groups of 19 are removed simultaneously with 90% aq. TFA, and the resulting product, without purification, is heated with (EtO) 3 CH in DMF at 90 0 C to give the bredinin 5'- phosphate derivative 20 in 47% yield from 19.
  • Each of the compounds is also preferred for use in preparation of medicaments for immunopotentiation, treating microbial and viral infections, particularly HCV, HIV, and HSV, and in treating biological conditions mediated therefrom.
  • Some of the Example compounds were screened and found to not be effective at a concentration of 20 ⁇ M or less using the assay described below. These compounds are also useful within the scope of the invention, since the invention is not meant to be limited to those compounds that are useful at a concentration of 20 ⁇ M or less.
  • Compounds may be useful as intermediates or prodrugs with undetectable activity in the present assay, or as final products that cause production of TNF- ⁇ at higher concentrations, such as 100 ⁇ M, 200 ⁇ M or 300 ⁇ M in the assays described herein. For example Loxoribine causes useful production of TNF-ce at 300 ⁇ M (see Pope et al. Cellular Immunology 162: 333-339 (1995)).
  • Candidate small molecule immuno-potentiators can be identified in vitro. Compounds are screened in vitro for their ability to activate immune cells and or ability to inhibit viral replication/activity. One marker of such activation is the induction of cytokine production, for example TNF-a production. Apoptosis inducing small molecules may be identified having this activity. These small molecule immuno-potentiators have potential utility as adjuvants and immuno-therapeutics.
  • the PBMCs are incubated for 18 h at 37°C in 5% CO 2 . Their ability to produce cytokines in response to the small molecule compounds is determined using a modified sandwich ELISA. [00524] Briefly supernatants from the PBMC cultures are assayed for secreted TNF using a primary plate bound antibody for capture followed by a secondary biotinylated anti-TNF antibody forming a sandwich. The biotinylated second antibody is then detected using streptavidin-Europium and the amount of bound europium is determined by time resolved fluorescence.
  • ILl-beta IL-12, IL-6, IFN-gamma, IL-10 etc.
  • ILl-beta IL-12, IL-6, IFN-gamma, IL-10 etc.
  • Compounds may be useful that cause production of TNF- ⁇ at higher concentrations, such as lOO ⁇ M, 200 ⁇ M or 300 ⁇ M in the assays described herein.
  • Loxoribine causes useful production of TNF- ⁇ at 300 ⁇ M (see Pope et al. Immunostimulatory Compound 7-Allyl-8-Oxoguanosine (Loxoribine) Induces a Distinct Subset of Murine Cytokines Cellular Immunology 162: 333-339 (1995)).
  • Blood is collected aseptically using a sterile collecting system consisting of a butterfly needle connected to a syringe (Becton Dickinson & Co., Rutherford, NJ).
  • Anticoagulation is obtained using sterile heparin (Elkins-Sinn Inc., Cherry Hill, NJ) (10 U/ml blood, final concentration). Heparin is chosen as anticoagulant rather than EDTA in whole blood experiments, since EDTA has been reported to inhibit cell function in bioassays and to inhibit the production of TNF. Incubation of heparinized whole blood in the absence of LPS does not result in detectable cytokine production. Screens are run as described above in the in vitro method.
  • TNF levels are expressed as nanograms per 10 9 monocytes, since monocyte counts change during administration of a particular 3,4-di(lH-indol-3-yl)- lH-pyrrole-2,5-dione, indolinone, chromen-4-one, derivatized pyridazine, staurosporine analog, nucleoside analog or other small molecule as described herein, and monocytes are the major source of TNF.
  • HCV Replicon RNA In Cell Lines (HCV Cell Based Assay) [00531] Cell lines, including Huh-11-7 or Huh 9-13, harboring HCV replicons (Lohmami, et al Science 285:110-113, 1999) are seeded at 5x10 cells/well in 96 well plates and fed media containing DMEM (high glucose), 10% fetal calf serum, penicillin-streptomycin and non-essential amino acids. Cells are incubated in a 5% CO 2 incubator at 37 0 C. At the end of the incubation period, total RNA is extracted and purified from cells using Qiagen Rneasy 96 Kit (Catalog No. 74182).
  • primers specific for HCV mediate both the reverse transcription (RT) of the HCV RNA and the amplification of the cDNA by polymerase chain reaction (PCR) using the TaqMan One-Step RT-PCR Master Mix Kit (Applied Biosystems catalog no. 4309169).
  • the nucleotide sequences of the RT-PCR primers which are located in the NS5B region of the HCV genome, are the following: [0100] HCV Forward primer "RBNS5bfor" [0101] 5 'GCTGCGGCCTGTCGAGCT: [0102] HCV Reverse primer "RBNS5Brev”: [0103] 5 'CAAGGTCGTCTCCGCATAC [0104] Detection of the RT-PCR product is accomplished using the Applied Biosystem (ABI) Prism 7700 Sequence Detection System (SDS) that detects the fluorescence that is emitted when the probe, which is labeled with a fluorescence reporter dye and a quencher dye, is processed during the PCR reaction.
  • SDS Sequence Detection System
  • the increase in the amount of fluorescence is measured during each cycle of PCR and reflects the increasing amount of RT-PCR product. Specifically, quantification is based on the threshold cycle, where the amplification plot crosses a defined fluorescence threshold. Comparison of the threshold cycles of the sample with a known standard provides a highly sensitive measure of relative template concentration in different samples (ABI User Bulletin #2 December 11, 1997). The data is analyzed using the ABI SDS program version 1.7. The relative template concentration can be converted to RNA copy numbers by employing a standard curve of HCV RNA standards with known copy number (ABI User Bulletin #2 December 11, 1997).
  • cells were seeded at 5x 103 cells/well in a 96 well plate and were incubated either with: 1) media containing 1% DMSO (0% inhibition control), 2) 100 international units, IU/ml Interferon-alpha 2b in media/1 %DMSO or 3) media/1 %DMSO containing a fixed concentration of compound.
  • 96 well plates as described above were then incubated at 37 0 C for 3 days (primary screening assay) or 4 days (IC 50 determination).
  • HIV-I Replication Assay The inhibitory effects of the nucleoside analogs on HIV-I replication may be due to the inhibition of virus-induced infectious focus formation in MAGI-CCR5 cells. Briefly, MAGI-CCR5 cells are seeded in a 96-well plate at 1.5 x 10 4 cells per well. The culture supernatants are removed on the next day, and fresh culture medium containing the virus (approximately 300 focus-forming units per well) and various concentrations of the test compounds are added to each well. On day 2 after viral infection, the culture supernatants are removed and fixing solution (1% formaldehyde and 0.2% glutaraldehyde in phosphate-buffered saline [PBS]) is added to each well.
  • fixing solution 1% formaldehyde and 0.2% glutaraldehyde in phosphate-buffered saline [PBS]
  • the cells are fixed at room temperature for 5 min and washed twice with PBS.
  • X-GaI staining solution (4 mM potassium ferrocyanide, 4 mM potassium ferricyanide, 2 mM magnesium chloride, and 0.4 mg of 5-bromo-4-chloro-3-indoyl- ⁇ -D-galactopyranoside per ml in PBS) is added to each well, and the cells are stained at 37°C for 45 min. The number of infected (blue) cells is counted microscopically.
  • ENV-Mediated Membrane Fusion Assay [00533] The inhibitory effects of the test compounds on HIV-I Env-mediated membrane fusion are determined by a P-D-galactosidase reporter gene system.
  • 293T cells are seeded in a six-well plate at 10 6 cells per well. The culture supernatants are removed on the next day, and the cells are transfected with 0.6 ⁇ g of Env expression vector, 0.2 ⁇ g of p-rev encoding HIV-I Rev, and 1.0 ⁇ g of pSV2tat encoding HIV-I Tat with Lipofectamine (Life Technologies).
  • the mixtures are removed and the cells are incubated with fresh culture medium for 2 days.
  • MAGI-CCR5 cells are seeded in a 96-well plate at 10 4 cells per well. Culture supernatants are removed on the next day, and fresh culture medium containing transfected 293T cells (10 4 cells per well) and various concentrations of the test compounds are added to each well.
  • the target and effector cell suspensions are incubated at 37 0 C.
  • Gal-Screen Tropix, Foster City, Calif.
  • the P- D-galactosidase activity in each well is measured with a luminometer (Microlumat LB96P; Berthold, Wildbad, Germany).
  • Pocket pets are assigned to groups of 7 animals; one group for each treatment and one to serve as a control (vehicle treated).
  • the compounds of the invention are formulated in water containing 5% Tween 80 (a polyoxyethylene sorbitan monooleate available from Aldrich Chemical Company, Inc., Milwaukee, Wis.).
  • Tween 80 a polyoxyethylene sorbitan monooleate available from Aldrich Chemical Company, Inc., Milwaukee, Wis.
  • the pocket pets are treated orally once daily for four consecutive days starting 24 hours after infection.
  • Antiviral activity is evaluated by comparing lesion development in compound treated versus vehicle treated pocket pets.
  • External lesions are scored 4, 7, 8 and 9 days after infection using the following scale: 0— no lesion, 1 —redness and swelling, 2--a few small vesicles, 3— several large vesicles, 4— large ulcers with necrosis and 5 ⁇ paralysis.
  • the maximum lesion score of each pocket pet is used to calculate the percentage lesion inhibition.
  • the percentage lesion inhibition is calculated as follows: 100 — Sum of maximum lesions scores of treatment group x 100 Sum of maximum lesion scores of vehicle group
  • Provisional Applications is incorporated herein by reference in their entirety: 60/599,717, filed August 5, 2004; 60/599,592, filed August 5, 2004; 60/600,850, filed August 11, 2004; 60/603,001, filed August 19,12004; 60,603,867, filed August 23, 2004; 60/612,070, filed September 21, 2004; 60/582,654), filed June 24, 2004; 60/614,963, filed September 21, 2004; and 60/590,459, filed July 22, 2004.
EP05791104A 2004-06-24 2005-06-24 Verbindungen als immunstimulatoren Withdrawn EP1765313A2 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP10179748A EP2277595A3 (de) 2004-06-24 2005-06-24 Verbindungen zur Immunopotenzierung

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US58265404P 2004-06-24 2004-06-24
US58809604P 2004-07-14 2004-07-14
US59045904P 2004-07-22 2004-07-22
US59959204P 2004-08-05 2004-08-05
US59971704P 2004-08-05 2004-08-05
US60085004P 2004-08-11 2004-08-11
US60300104P 2004-08-19 2004-08-19
US60386704P 2004-08-23 2004-08-23
US61207004P 2004-09-21 2004-09-21
US61496304P 2004-09-30 2004-09-30
PCT/US2005/022769 WO2006002422A2 (en) 2004-06-24 2005-06-24 Compounds for immunopotentiation

Publications (1)

Publication Number Publication Date
EP1765313A2 true EP1765313A2 (de) 2007-03-28

Family

ID=35601821

Family Applications (2)

Application Number Title Priority Date Filing Date
EP05791104A Withdrawn EP1765313A2 (de) 2004-06-24 2005-06-24 Verbindungen als immunstimulatoren
EP10179748A Withdrawn EP2277595A3 (de) 2004-06-24 2005-06-24 Verbindungen zur Immunopotenzierung

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP10179748A Withdrawn EP2277595A3 (de) 2004-06-24 2005-06-24 Verbindungen zur Immunopotenzierung

Country Status (4)

Country Link
EP (2) EP1765313A2 (de)
JP (2) JP2008504292A (de)
CA (1) CA2571421A1 (de)
WO (1) WO2006002422A2 (de)

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008504286A (ja) * 2004-06-24 2008-02-14 カイロン コーポレイション 小分子免疫増強因子およびその検出のためのアッセイ
CN1947717B (zh) * 2005-10-14 2012-09-26 卓敏 选择性抑制腺苷酸环化酶1的化合物在制备用于治疗神经性疼痛和炎性疼痛的药物中的应用
AU2006310337B9 (en) 2005-11-04 2013-11-28 Novartis Ag Adjuvanted influenza vaccines including cytokine-inducing agents
NZ567978A (en) 2005-11-04 2011-09-30 Novartis Vaccines & Diagnostic Influenza vaccine with reduced amount of oil-in-water emulsion as adjuvant
AU2006310336B2 (en) 2005-11-04 2011-02-03 Seqirus UK Limited Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
US20090291955A1 (en) * 2005-11-15 2009-11-26 Crane Sheldon N Azacyclohexane Derivatives as Inhibitors of Stearoyl-Coenzyme a Delta-9 Desaturase
EP1959945B1 (de) * 2005-12-13 2014-12-03 McGill University Fenretinid zur korrektur des lipidungleichgewichts bei einer person
JP2009520730A (ja) * 2005-12-23 2009-05-28 ヤド・テヒノロギース・ゲーエムベーハー アレルギー性疾患を治療および予防するための手段および方法
BR122019013216B8 (pt) 2006-01-27 2021-07-27 Seqirus Uk Ltd ensaio imunológico para análise de uma vacina contra influenza
EP2382987A1 (de) 2006-03-24 2011-11-02 Novartis Vaccines and Diagnostics GmbH Lagerung von Influenza-Impfstoffen ohne Kühlung
CA2647942A1 (en) 2006-03-31 2007-11-08 Novartis Ag Combined mucosal and parenteral immunization against hiv
GB0614460D0 (en) 2006-07-20 2006-08-30 Novartis Ag Vaccines
US8158810B2 (en) 2006-07-27 2012-04-17 Gilead Sciences, Inc. ALDH-2 inhibitors in the treatment of addiction
US20080032995A1 (en) * 2006-07-27 2008-02-07 Jeff Zablocki Aldh-2 inhibitors in the treatment of drug addiction
CA2659552A1 (en) 2006-08-16 2008-02-21 Novartis Ag Immunogens from uropathogenic escherichia coli
WO2008024439A2 (en) * 2006-08-22 2008-02-28 Concert Pharmaceuticals Inc. 4-aminoquinazoline derivatives and methods of use thereof
WO2008032219A2 (en) 2006-09-11 2008-03-20 Novartis Ag Making influenza virus vaccines without using eggs
EP2077842A2 (de) * 2006-10-31 2009-07-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Verwendung eines egfr-antagonisten zur behandlung von glomerolonephritis
US20110045022A1 (en) 2006-12-06 2011-02-24 Theodore Tsai Vaccines including antigen from four strains of influenza virus
GB0700562D0 (en) 2007-01-11 2007-02-21 Novartis Vaccines & Diagnostic Modified Saccharides
WO2008141455A1 (en) * 2007-05-23 2008-11-27 Merck Frosst Canada Ltd. Bicyclic heteroaromatic compounds as inhibitors of stearoyl-coenzyme a delta-9 desaturase
BRPI0813866A2 (pt) 2007-06-27 2015-01-06 Novartis Ag Vacinas contra influenza com baixo teor de aditivos
GB0713880D0 (en) 2007-07-17 2007-08-29 Novartis Ag Conjugate purification
GB0810305D0 (en) 2008-06-05 2008-07-09 Novartis Ag Influenza vaccination
CA2712750A1 (en) * 2008-01-24 2009-07-30 Gilead Palo Alto, Inc. Aldh-2 inhibitors in the treatment of addiction
ES2326459B1 (es) * 2008-04-08 2010-05-28 Universidad De Oviedo Indolocarbazoles glicosilados, su procedimiento de obtencion y sus usos.
EP2158912A1 (de) 2008-08-25 2010-03-03 Ratiopharm GmbH Pharmazeutische Zusammensetzung mit N-[3-chlor-4-(3-fluorophenyl)methoxy)phenyl)6-(5(((2-(methylsulfonyl)ethyl)amino)methyl)-2-furyl)-4-chinazolinamin
EP2158913A1 (de) 2008-08-25 2010-03-03 Ratiopharm GmbH Pharmazeutische Zusammensetzung mit N-[3-chlor-4-(3-fluorophenyl)methoxy)phenyl)6-(5(((2-(methylsulfonyl)ethyl)amino)methyl)-2-furyl)-4-chinazolinamin
CA2737948A1 (en) * 2008-09-26 2010-04-01 Inserm (Institut National De La Sante Et De La Recherche Medicale) Host cell kinases as targets for antiviral therapies against hcv infection
US8809343B2 (en) 2008-12-26 2014-08-19 Fudan University Pyrimidine derivative, preparation method and use thereof
HRP20220756T1 (hr) 2009-07-15 2022-09-02 Glaxosmithkline Biologicals S.A. Proteinski pripravci rsv f i postupci za izradu istih
EP2560652A4 (de) * 2010-04-23 2013-08-14 Kineta Inc Antivirale verbindungen
WO2012064981A2 (en) * 2010-11-10 2012-05-18 National Jewish Health Methods to test allergic conditions
CN102552271B (zh) * 2010-12-09 2014-08-06 中国科学院上海药物研究所 一类喹唑啉类化合物在制备抗黄病毒科病毒的药物中的用途
EP2667892B1 (de) 2011-01-26 2019-03-27 GlaxoSmithKline Biologicals SA Rsv-immunisierungstherapie
EP2678019A4 (de) * 2011-02-25 2015-05-13 Kineta Inc Verfahren und zellen zur identifizierung von rig-i-pfadregulatoren
SG194755A1 (en) 2011-05-13 2013-12-30 Novartis Ag Pre-fusion rsv f antigens
WO2014170910A1 (en) 2013-04-04 2014-10-23 Natco Pharma Limited Process for the preparation of lapatinib
WO2015009889A1 (en) 2013-07-18 2015-01-22 Concert Pharmaceuticals, Inc. Deuterated intedanib derivatives and their use for the treatment of proliferative disorders
WO2016046734A2 (en) * 2014-09-22 2016-03-31 University Of The Western Cape Compounds and compositions for treatment of tuberculosis
DE202014010366U1 (de) 2014-11-24 2015-05-15 Clariant International Ltd. Verwendung von Polyhydroxyfettsäureamiden zur Verbesserung der Nassabriebbeständigkeit von wässrigen Dispersionen
DE102014017368A1 (de) 2014-11-24 2015-05-28 Clariant International Ltd. Verwendung von Polyhydroxyfettsäureamiden zur Verbesserung der Nassabriebbeständigkeit von wässrigen Dispersionsfarben
CN106478598B (zh) * 2016-08-30 2018-11-13 山东罗欣药业集团股份有限公司 一种凡德他尼水合物晶体及其制备方法
CN106397401B (zh) * 2016-08-30 2018-11-13 山东罗欣药业集团股份有限公司 一种抗癌药物的晶体化合物及其制备方法
CN109223793B (zh) * 2018-08-24 2020-07-28 江苏理工学院 一种化合物c1作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN109223796B (zh) * 2018-08-24 2020-07-28 江苏理工学院 一种化合物b6作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN109223795B (zh) * 2018-08-24 2020-07-28 江苏理工学院 一种化合物b2作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN108853110B (zh) * 2018-08-24 2020-07-28 江苏理工学院 一种化合物a8作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN109223794B (zh) * 2018-08-24 2020-07-28 江苏理工学院 一种化合物c6作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN109053544B (zh) * 2018-08-24 2020-07-28 江苏理工学院 一种化合物b11作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN109232573A (zh) * 2018-08-24 2019-01-18 江苏理工学院 一种化合物b1作为组蛋白甲基转移酶nsd3活性抑制剂及其应用
CN109400567A (zh) * 2018-12-05 2019-03-01 华中药业股份有限公司 一种合成甘草素的方法
EP3725788A1 (de) * 2019-04-15 2020-10-21 Bayer AG Neuartige heteroarylsubstituierte aminoalkyl-azol-verbindungen als pestizide
CN112300228A (zh) * 2020-10-31 2021-02-02 青岛姿之妍化妆品科技有限责任公司 一种包括含有根皮三酚基团的根皮素衍生物的化妆品及其制备方法
CN112125940A (zh) * 2020-10-31 2020-12-25 青岛姿之妍化妆品科技有限责任公司 含有四乙酰葡萄糖苷改性的根皮素的化妆品及其制备方法

Family Cites Families (121)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE567431A (de) 1957-05-07
US4323581A (en) 1978-07-31 1982-04-06 Johnson & Johnson Method of treating carcinogenesis
IE52670B1 (en) 1981-03-03 1988-01-20 Leo Ab Heterocyclic carboxamides,compositions containing such compounds,and processes for their preparation
IL73534A (en) 1983-11-18 1990-12-23 Riker Laboratories Inc 1h-imidazo(4,5-c)quinoline-4-amines,their preparation and pharmaceutical compositions containing certain such compounds
US4806352A (en) 1986-04-15 1989-02-21 Ribi Immunochem Research Inc. Immunological lipid emulsion adjuvant
ES2037072T3 (es) 1986-12-19 1993-06-16 Duphar International Research B.V Suspension coadyuvante estabilizada que comprende bromuro de dimetil-dioctadecil-amonio.
EP0303697B1 (de) 1987-03-09 1997-10-01 Kyowa Hakko Kogyo Co., Ltd. Derivate des physiologisch aktiven mittels k-252
EP0283085B1 (de) 1987-03-17 1992-11-11 Akzo N.V. Adjuvanzienmischung
EP0383919A4 (en) 1988-02-04 1992-08-05 Kyowa Hakko Kogyo Co., Ltd. Staurosporin derivatives
SK75289A3 (en) 1988-02-10 1998-05-06 Hoffmann La Roche Substituted pyrroles, their use for producing a drug, and the drug on their base
DE3841091A1 (de) 1988-12-07 1990-06-13 Behringwerke Ag Synthetische antigene, verfahren zu ihrer herstellung und ihre verwendung
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
ES2055785T3 (es) 1989-01-17 1994-09-01 Eniricerche Spa Peptidos sinteticos y su uso como vehiculos universales para la preparacion de conjugados inmunogenos aptos para el desarrollo de vacunas sinteticas.
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
CA2011896C (en) 1989-04-21 2001-05-08 Mark Werner Ringworm vaccine
EP0474727B1 (de) 1989-05-19 1997-07-23 Genentech, Inc. Her2 extrazellulare domäne
HU212924B (en) 1989-05-25 1996-12-30 Chiron Corp Adjuvant formulation comprising a submicron oil droplet emulsion
JP3859695B2 (ja) 1989-08-04 2006-12-20 バーレックス ラボラトリーズ インコーポレイテッド C―erbB―2外部ドメイン:GP75
US5439936A (en) 1989-10-03 1995-08-08 The Regents Of The University Of California Method of treating certain tumors using illudin analogs
IT1237764B (it) 1989-11-10 1993-06-17 Eniricerche Spa Peptidi sintetici utili come carriers universali per la preparazione di coniugati immunogenici e loro impiego per lo sviluppo di vaccini sintetici.
ATE128628T1 (de) 1990-08-13 1995-10-15 American Cyanamid Co Faser-hemagglutinin von bordetella pertussis als träger für konjugierten impfstoff.
US5153312A (en) 1990-09-28 1992-10-06 American Cyanamid Company Oligosaccharide conjugate vaccines
US5284652A (en) 1990-10-26 1994-02-08 Pier Allan C Dermatophyte vaccine
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
US5268376A (en) 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
WO1993007153A1 (en) 1991-10-10 1993-04-15 Schering Corporation 4'-(n-substituted-n-oxide)staurosporine derivatives
US5266575A (en) 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
AU3064992A (en) 1991-11-08 1993-06-07 University Of Southern California Compositions containing k-252 compounds for potentiation of neurotrophin activity
CA2117303C (en) 1991-11-29 2000-10-17 Sunil Chada Anti-cancer immunotherapeutic vector constructs
AU661533B2 (en) 1992-01-20 1995-07-27 Astrazeneca Ab Quinazoline derivatives
DE69333110T2 (de) 1992-03-02 2004-05-06 Chiron S.P.A. Mit dem cytotoxin von helicobacter pylori assoziiertes, immunodominantes antigen verwendbar in impfstoffen und zur diagnose
IT1262896B (it) 1992-03-06 1996-07-22 Composti coniugati formati da proteine heat shock (hsp) e oligo-poli- saccaridi, loro uso per la produzione di vaccini.
US5756494A (en) 1992-07-24 1998-05-26 Cephalon, Inc. Protein kinase inhibitors for treatment of neurological disorders
US5621100A (en) 1992-07-24 1997-04-15 Cephalon, Inc. K-252a derivatives for treatment of neurological disorders
CA2123895A1 (en) 1992-09-21 1994-03-31 Tatsuya Tamaoki A therapeutic agent for thrombocytopenia
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
US5801005A (en) 1993-03-17 1998-09-01 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis of malignancies in which the HER-2/neu oncogene is associated
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
KR100341341B1 (ko) 1993-07-15 2002-11-25 미네소타 마이닝 앤드 매뉴팩춰링 캄파니 이미다조[4,5-c]피리딘-4-아민
US5624949A (en) 1993-12-07 1997-04-29 Eli Lilly And Company Protein kinase C inhibitors
AU1339895A (en) 1993-12-23 1995-07-10 Eli Lilly And Company Protein kinase c inhibitors
US6083903A (en) 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
US5482936A (en) 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
IL117483A (en) 1995-03-17 2008-03-20 Bernard Brodeur MENINGITIDIS NEISSERIA shell protein is resistant to proteinase K.
ES2332984T3 (es) 1995-03-30 2010-02-16 Pfizer Products Inc. Derivados de quinazolinas.
GB9508538D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
US6331555B1 (en) 1995-06-01 2001-12-18 University Of California Treatment of platelet derived growth factor related disorders such as cancers
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5880141A (en) 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
GB9513261D0 (en) 1995-06-29 1995-09-06 Smithkline Beecham Biolog Vaccines
US5554519A (en) 1995-08-07 1996-09-10 Fermalogic, Inc. Process of preparing genistein
AU6888196A (en) 1995-08-11 1997-03-12 Yale University Glycosylated indolocarbazole synthesis
FR2741881B1 (fr) 1995-12-01 1999-07-30 Centre Nat Rech Scient Nouveaux derives de purine possedant notamment des prorietes anti-proliferatives et leurs applications biologiques
IL123568A (en) 1995-12-08 2001-08-08 Janssen Pharmaceutica Nv History (Imidazole - 5Il) Methyl-2-quinolinone inhibitors of Prenzyl transferase, their preparation, pharmaceutical preparations containing them and their preparation and drugs containing them
EP0907738A1 (de) 1996-04-02 1999-04-14 Smithkline Beecham Corporation Neue verbindungen
US5849733A (en) 1996-05-10 1998-12-15 Bristol-Myers Squibb Co. 2-thio or 2-oxo flavopiridol analogs
JP2000508178A (ja) 1996-05-14 2000-07-04 スミスクライン・ビーチャム・コーポレイション 新規化合物
KR100447918B1 (ko) 1996-07-25 2005-09-28 동아제약주식회사 대장을포함한위장관보호작용을갖는플라본및플라바논화합물
DE19630390A1 (de) 1996-07-26 1998-01-29 Chiron Behring Gmbh & Co Proteine, insbesondere Membranproteine von Helicobacter pylori, ihre Herstellung und Verwendung
CA2264496C (en) 1996-09-04 2008-10-07 Takara Shuzo Co., Ltd. Fungal antigens and process for producing the same
US5908934A (en) 1996-09-26 1999-06-01 Bristol-Myers Squibb Company Process for the preparation of chiral ketone intermediates useful for the preparation of flavopiridol and analogs
ES2241042T3 (es) 1996-10-11 2005-10-16 The Regents Of The University Of California Conjugados de polinucleotido inmunoestimulador/ molecula inmunomoduladora.
EP1400592A1 (de) 1996-10-31 2004-03-24 Human Genome Sciences, Inc. Polynukleotide und Sequenzen aus Streptococcus Pneumoniae
CO4950519A1 (es) 1997-02-13 2000-09-01 Novartis Ag Ftalazinas, preparaciones farmaceuticas que las comprenden y proceso para su preparacion
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6299881B1 (en) 1997-03-24 2001-10-09 Henry M. Jackson Foundation For The Advancement Of Military Medicine Uronium salts for activating hydroxyls, carboxyls, and polysaccharides, and conjugate vaccines, immunogens, and other useful immunological reagents produced using uronium salts
ATE432348T1 (de) 1997-06-06 2009-06-15 Univ California Inhibitoren von immunstimulatorischen dna sequenz aktivität
GB9712347D0 (en) 1997-06-14 1997-08-13 Smithkline Beecham Biolog Vaccine
GB9713156D0 (en) 1997-06-20 1997-08-27 Microbiological Res Authority Vaccines
US6800744B1 (en) 1997-07-02 2004-10-05 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
EP1017394B1 (de) 1997-07-12 2005-12-07 Cancer Research Technology Limited Cyclin-abhängige-kinase inhibierende purinderivate
US5948413A (en) 1997-07-17 1999-09-07 Board Of Trustees Operating Michigan State University Method and vaccine for treatment of pythiosis insidiosi in humans and lower animals
WO1999011241A1 (en) 1997-09-05 1999-03-11 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins
ATE352624T1 (de) 1997-11-21 2007-02-15 Serono Genetics Inst Sa Chlamydia pneumoniae genomische sequenzen und polypeptiden, fragmenten und anwendungen davon für nachweis, prevention und heilung
BR9814912A (pt) 1997-11-28 2000-10-03 Genset Sa Sequência genÈmica e polipeptìdeos de chlamydia trachomatis, fragmentos dos mesmos e usos dos mesmos, em particular para o diagnóstico, a prevenção e o tratamento de infecção
UA67760C2 (uk) 1997-12-11 2004-07-15 Міннесота Майнінг Енд Мануфакчурінг Компані Імідазонафтиридин та тетрагідроімідазонафтиридин, фармацевтична композиція, спосіб індукування біосинтезу цитокінів та спосіб лікування вірусної інфекції, проміжні сполуки
GB9800569D0 (en) * 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
US6303114B1 (en) 1998-03-05 2001-10-16 The Medical College Of Ohio IL-12 enhancement of immune responses to T-independent antigens
GB9807721D0 (en) 1998-04-08 1998-06-10 Chiron Spa Antigen
PL354714A1 (en) 1998-04-09 2004-02-09 Smithkline Beecham Biologicals S.A. Adjuvant compositions
GB9810193D0 (en) 1998-05-12 1998-07-08 Smithkline Beecham Biolog Novel compounds
CA2314156C (en) 1998-05-29 2010-05-25 Sugen, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
DE69936285D1 (de) 1998-09-30 2007-07-26 Us Army INDOLOi2,1-BöQUINAZOLIN-6,12-DIONE ANTIMALARIA DERIVATE UND METHODEN ZUR BEHANDLUNG VON MALARIA
AU1202200A (en) 1998-10-09 2000-05-01 Chiron Corporation Neisseria genomic sequences and methods of their use
EP1588714A2 (de) 1998-10-16 2005-10-26 GlaxoSmithKline Biologicals S.A. Adjuvanzsysteme und Impfstoffe
US20030083242A1 (en) 1998-11-06 2003-05-01 Alphonse Galdes Methods and compositions for treating or preventing peripheral neuropathies
JP2002529069A (ja) 1998-11-12 2002-09-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア クラミジア・ニューモニエのゲノム配列
GB9828000D0 (en) 1998-12-18 1999-02-10 Chiron Spa Antigens
DE60032120T2 (de) 1999-03-19 2007-09-20 Glaxosmithkline Biologicals S.A. Impstoff gegen Streptococcus pneumoniae
KR100922031B1 (ko) 1999-04-19 2009-10-19 글락소스미스클라인 바이오로지칼즈 에스.에이. 백신
WO2000067161A2 (en) 1999-05-04 2000-11-09 Grant Lee H Method and apparatus for categorizing and retrieving network pages and sites
WO2001004125A1 (fr) 1999-07-13 2001-01-18 Kyowa Hakko Kogyo Co., Ltd. Derives de staurosporine
AU6620300A (en) 1999-08-03 2001-02-19 Ohio State University, The Polypeptides and polynucleotides for enhancing immune reactivity to her-2 protein
CA2383413A1 (en) 1999-09-24 2001-03-29 Smithkline Beecham Biologicals S.A. Use of combination of polyoxyethylene sorbitan ester and octoxynol as adjuvant and its use in vaccines
JP2003509452A (ja) 1999-09-24 2003-03-11 スミスクライン ビーチャム バイオロジカルズ ソシエテ アノニム ポリオキシエチレンアルキルエーテル又はエステル及び少なくとも一つのノニオン界面活性剤を含有するアジュバント
SK287401B6 (sk) * 1999-11-05 2010-09-07 Astrazeneca Ab Deriváty chinazolínu, spôsob ich prípravy, farmaceutická kompozícia, ktorá ich obsahuje, a ich použitie
EP2281571A3 (de) 2000-01-17 2012-04-25 Novartis Vaccines and Diagnostics S.r.l. Membranvesikel (omv) impfstoff, der n. meningitidis serogruppe b membranproteine enthält
AT409085B (de) 2000-01-28 2002-05-27 Cistem Biotechnologies Gmbh Pharmazeutische zusammensetzung zur immunmodulation und herstellung von vakzinen
NZ520640A (en) 2000-02-15 2005-04-29 Upjohn Co Pyrrole substituted 2-indolinone protein kinase inhibitors
AU2001253427B2 (en) 2000-04-12 2007-02-08 Genaera Corporation A process for the preparation of 7.alpha.-hydroxy 3-aminosubstituted sterols using intermediates with an unprotected 7.alpha.-hydroxy group
CZ299561B6 (cs) 2000-06-30 2008-09-03 Glaxo Group Limited Chinazolinaminový derivát a farmaceutický prostredek
NZ524461A (en) * 2000-08-18 2004-12-24 Millennium Pharm Inc Quinazoline derivatives as kinase inhibitors
AU8743001A (en) 2000-08-28 2002-03-13 Aventis Pasteur Moraxella polypeptides and corresponding dna fragments and uses thereof
US6677450B2 (en) 2000-10-06 2004-01-13 Bristol-Myers Squibb Company Topoisomerase inhibitors
EP2284181A1 (de) 2000-10-27 2011-02-16 Novartis Vaccines and Diagnostics S.r.l. Nukleinsäure und Proteine von Gruppen Streptokokkengruppen A und B
WO2002062826A1 (fr) 2001-02-07 2002-08-15 Vadim Viktorovich Novikov Procede de fabrication des peptides
US7407664B2 (en) 2001-05-18 2008-08-05 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Peptide vaccines against group A streptococci
WO2003004505A1 (en) 2001-07-02 2003-01-16 Debiopharm S.A. Oxaliplatin active substance with a very low content of oxalic acid
KR100484504B1 (ko) 2001-09-18 2005-04-20 학교법인 포항공과대학교 쿠커비투릴 유도체를 주인 분자로서 포함하고 있는 내포화합물 및 이를 포함한 약제학적 조성물
US20030134846A1 (en) 2001-10-09 2003-07-17 Schering Corporation Treatment of trypanosoma brucei with farnesyl protein transferase inhibitors
GB0126879D0 (en) * 2001-11-08 2002-01-02 Astrazeneca Ab Combination therapy
US6927036B2 (en) 2002-02-19 2005-08-09 Xero Port, Inc. Methods for synthesis of prodrugs from 1-acyl-alkyl derivatives and compositions thereof
GB0210128D0 (en) 2002-05-02 2002-06-12 Chiron Spa Nucleic acids and proteins from streptococcus groups A & B
US6900342B2 (en) 2002-05-10 2005-05-31 Dabur India Limited Anticancer taxanes such as paclitaxel, docetaxel and their structural analogs, and a method for the preparation thereof
US6727272B1 (en) 2002-07-15 2004-04-27 Unitech Pharmaceuticals, Inc. Leflunomide analogs for treating rheumatoid arthritis
US20040047835A1 (en) * 2002-09-06 2004-03-11 Cell Therapeutics, Inc. Combinatorial drug therapy using polymer drug conjugates
ES2504166T3 (es) 2002-09-13 2014-10-08 Novartis Vaccines And Diagnostics, Inc. Vacuna de estreptococo del grupo B
US20060067947A1 (en) * 2002-09-19 2006-03-30 Orient Cancer Therapy Co., Ltd. Immunotherapeutic for cancer
MXPA05013260A (es) 2003-06-26 2006-03-09 Chiron Corp Composiciones inmunogenicas para chlamydia trachomatis.
PT1648500E (pt) 2003-07-31 2014-10-10 Novartis Vaccines & Diagnostic Composições imunogénicas para estreptococos piogenes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006002422A2 *

Also Published As

Publication number Publication date
WO2006002422A2 (en) 2006-01-05
WO2006002422A9 (en) 2006-09-21
EP2277595A3 (de) 2011-09-28
JP2008504292A (ja) 2008-02-14
EP2277595A2 (de) 2011-01-26
WO2006002422A3 (en) 2006-07-13
CA2571421A1 (en) 2006-01-05
JP2012184264A (ja) 2012-09-27

Similar Documents

Publication Publication Date Title
WO2006002422A2 (en) Compounds for immunopotentiation
US20100226931A1 (en) Compounds for immunopotentiation
JP4769810B2 (ja) イミダゾキノリン化合物
US8063063B2 (en) Immunopotentiating compounds
US20110104186A1 (en) Small molecule immunopotentiators and assays for their detection
US8173657B2 (en) Imidazoquinoxaline compounds as immunomodulators
JP3167788B2 (ja) 肝炎の処置に有用な1,3−オキサチオラン
CN106459063A (zh) Mertk‑特异性嘧啶化合物
CN105722831B (zh) 哒嗪酮化合物及其用途
JP2018536681A (ja) インフルエンザウイルス複製の阻害剤、その適用方法および使用
JP2003512375A (ja) Hiv逆転写酵素阻害剤としての縮合ナフチリジン
TW201249796A (en) Heterocyclic modulators of lipid synthesis
CN102271681A (zh) Cxcr7调节剂
JP2022025116A (ja) 抗菌性化合物
AU2017356214A1 (en) Heterocyclic modulators of lipid synthesis
WO2004094424A1 (ja) 含窒素複素環化合物およびその用途
AU2012209025A1 (en) Imidazoquinoline compounds
JP2012107074A (ja) 小分子免疫増強因子およびその検出のためのアッセイ

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070105

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20080124

RIN1 Information on inventor provided before grant (corrected)

Inventor name: XU, FENGM/S R338

Inventor name: VALIANTE, NICHOLASM/S R338

Inventor name: SILVER, JOEL B.

RIN1 Information on inventor provided before grant (corrected)

Inventor name: XU, FENG

Inventor name: VALIANTE, NICHOLAS

Inventor name: SILVER, JOEL B.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20150106