US20130190395A1 - Autoimmune disorder treatment using rxr agonists - Google Patents

Autoimmune disorder treatment using rxr agonists Download PDF

Info

Publication number
US20130190395A1
US20130190395A1 US13/714,051 US201213714051A US2013190395A1 US 20130190395 A1 US20130190395 A1 US 20130190395A1 US 201213714051 A US201213714051 A US 201213714051A US 2013190395 A1 US2013190395 A1 US 2013190395A1
Authority
US
United States
Prior art keywords
carbons
rxr agonist
alkyl
rxr
lower alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/714,051
Other languages
English (en)
Inventor
Roshantha A. Chandraratna
Ethan Dmitrovsky
Elizabeth Nowak
Randolph Noelle
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.)
Dartmouth College
IO Therapeutics Inc
Original Assignee
Dartmouth College
IO Therapeutics 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
Priority to US13/714,051 priority Critical patent/US20130190395A1/en
Application filed by Dartmouth College, IO Therapeutics Inc filed Critical Dartmouth College
Assigned to DARTMOUTH COLLEGE reassignment DARTMOUTH COLLEGE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOELLE, RANDOLPH J., DMITROVSKY, ETHAN, NOWAK, ELIZABETH
Assigned to Io Therapeutics, Inc. reassignment Io Therapeutics, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANDRARATNA, ROSHANTHA A.
Assigned to Io Therapeutics, Inc. reassignment Io Therapeutics, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANDRARATNA, ROSHANTHA A.
Assigned to TRUSTEES OF DARTMOUTH COLLEGE reassignment TRUSTEES OF DARTMOUTH COLLEGE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOELLE, RANDOLPH, NOWAK, ELIZABETH, DMITROVSKY, ETHAN
Publication of US20130190395A1 publication Critical patent/US20130190395A1/en
Priority to US14/507,730 priority patent/US10653650B2/en
Priority to US14/626,339 priority patent/US10034845B2/en
Priority to US14/732,328 priority patent/US20150342917A1/en
Priority to US15/341,969 priority patent/US10285960B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: DARTMOUTH COLLEGE
Priority to US15/852,580 priority patent/US10201512B2/en
Priority to US15/988,965 priority patent/US10695307B2/en
Priority to US16/228,217 priority patent/US10945976B2/en
Priority to US16/736,705 priority patent/US20200163915A1/en
Priority to US16/742,600 priority patent/US20200155488A1/en
Priority to US16/742,616 priority patent/US11547684B2/en
Priority to US17/126,714 priority patent/US11166927B2/en
Priority to US17/126,787 priority patent/US11246845B2/en
Priority to US17/560,035 priority patent/US11576881B2/en
Priority to US17/586,657 priority patent/US11793781B2/en
Priority to US17/859,743 priority patent/US20220370386A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/201Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having one or two double bonds, e.g. oleic, linoleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47042-Quinolinones, e.g. carbostyril
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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

  • autoimmune disorders Attempts to treat autoimmune disorders have met with limited success. This is due, in part, to the fact that the etiology of autoimmune disorders is a complex response based in part on a combination of factors, including, without limitation, genetic make-up of individual, gender or hormonal status, bacterial or viral infection, metal or chemical toxin exposure, vaccinations or immunizations, stress, trauma, smoking and/or nutritional deficiencies. Therefore, compounds, compositions, and methods that can reduce a symptom associated with an autoimmune disorder, inflammation associated with an autoimmune disorder, and/or a transplant rejection would be highly desirable.
  • Na ⁇ ve CD4 + T cells play a central role in immune protection. They do so through their capacity to help B cells make antibodies, to induce macrophages to develop enhanced microbicidal activity, to recruit neutrophils, eosinophils, and basophils to sites of infection and inflammation, and, through their production of cytokines and chemokines, to orchestrate the full panoply of immune responses.
  • Na ⁇ ve CD4 + T cells are multipotential precursors that differentiate into various T cell subsets, such as, e.g., T helper (Th) cells (also called T effector cells) and T regulatory (Treg) cells. T helper cells are characterized by their distinct functions and include Th1, Th2, and Th17.
  • Th1 cells aid in the clearance of intracellular bacteria and viruses, secrete IFN- ⁇ in response to the cytokine interleukin-12 (IL-12), and require the transcription factors T-box21 (T-bet) and signal transducer and activator of transcription 1 (Stat1) and (Stat4).
  • Th2 cells help control extracellular pathogens, secrete the cytokines IL-4, IL-5 and IL-13, and require transcription factors GATA-binding protein 3 (GATA-3) and Stat6.
  • Th17 cells provide protection against fungi and various other extracellular bacteria, secrete the pro-inflammatory cytokine IL-17A, and express the transcription factor retinoic acid orphan receptor gamma (ROR ⁇ t).
  • Treg cells play a critical role in maintaining self-tolerance as well as in regulating immune responses and express the transcription factor forkhead box P3 (FoxP3).
  • Tregs normally develop in the thymus, but can also differentiate from na ⁇ ve CD4 + cells stimulated with TGF- ⁇ and IL-2. Development and differentiation of Treg cells, as well as expression of FoxP3, require the transcription factor Stat5.
  • Th17 plays a key role in the pathogenesis of autoimmune diseases and protection against bacterial infections, while Treg functions to restrain excessive helper T-cell responses.
  • immunosuppressive Tregs cells and pro-inflammatory Th17 cells functionally antagonize each other.
  • Th17 and Treg cells may be crucial for the stability of immune homeostasis. Once the equilibrium is broken, the destabilization may lead to chronic inflammation and autoimmunity.
  • dysregulation or overproduction of IL-6 leads to autoimmune diseases such as multiple sclerosis (MS) and rheumatoid arthritis (RA), in which Th17 cells are considered to be the primary cause of pathology.
  • MS multiple sclerosis
  • RA rheumatoid arthritis
  • Th17 cells are considered to be the primary cause of pathology.
  • Clinical evidence indicates that both defects in Treg function or reduced numbers, as well as Th17 activity are important in several autoimmune diseases, including seronegative arthritis in adults, and childhood arthritis (juvenile idiopathic arthritis). Therefore, an effective approach in the treatment of various autoimmune and inflammatory diseases will be to normalize the balance between Treg and Th17 cell development.
  • RARs Retinoic Acid Receptors
  • RXRs Retinoid X Receptors
  • Retinoic acid the physiological hormone of all three RARs, has been shown to enhance the in vitro differentiation of Treg cells that suppress immunity.
  • RA can also inhibit the differentiation of pro-inflammatory Th17 cells that have been casually implicated in the development of many human autoimmune diseases.
  • RAR agonists Based on this ability to restore a normal Th17/Treg cell ratio by decreasing Th17 cells while simultaneously increasing Treg cells, RAR agonists have been proposed as effective therapeutic compounds for the treatment of inflammatory and autoimmune disorders.
  • recent findings have identified retinoid signaling through RARs as being required for the initial development of Th17 cell mediated immune responses and inflammation. These counteracting effects of RAR pan agonists on Th17 cell development bring into question the value of such compounds as anti-inflammatory and immunosuppressive agents.
  • RAR agonists like RA have been used to treat autoimmune disorders associated with inflammation, their usefulness in clinical practice has been limited due to unwanted side effects and counter-therapeutic inflammatory effects.
  • compounds and compositions that maintain the ability to inhibit Th17 cell formation and function and to promote Treg cell formation, but not possess any pro-inflammatory activities and other unwanted side effects associated with RAR pan agonists like RA.
  • Such compounds will be of considerable therapeutic value as immunomodulatory agents.
  • the present specification discloses compounds, compositions, and methods for treating an individual suffering from an autoimmune disorder. This is accomplished by administering a therapeutically effective amount of a RXR agonist or composition comprising such agonist to an individual suffering from an autoimmune disorder.
  • a RXR agonist or composition comprising such agonist to an individual suffering from an autoimmune disorder.
  • the disclosed RXR agonists can control the Th17/Treg cell number ratio by elevating Treg cell numbers and suppressing Th17 cell numbers.
  • the disclosed RXR agonists would be useful in treating an autoimmune disorder.
  • RXR agonist examples include a compound having the structure of formula I,
  • Z is a radical having the structure of Formula II:
  • Y is cycloalkyl or cycloalkenyl of 3 to 8 carbons optionally substituted with one or two R 4 groups, or Y is selected from phenyl, pyridyl, thienyl, furyl, pyrrolyl, pyridazinyl, pyrimidiyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen, and
  • B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —
  • aspects of the present specification disclose a method of treating an autoimmune disorder, the method comprising the step of administering to an individual in need thereof a therapeutically effective amount of a RXR agonist, wherein administration of the compound or composition reduces a symptom associated with the autoimmune disorder, thereby treating the individual.
  • aspects of the present specification also disclose a use of a RXR agonist to treat an autoimmune disorder, wherein administration of the compound or composition reduces a symptom associated with the autoimmune disorder, thereby treating the individual.
  • Non-limiting examples of a RXR agonist include a compound or a composition disclosed herein.
  • the autoimmune disorder can be a systemic autoimmune disorder or an organ-specific autoimmune disorder.
  • Non-limiting examples of an autoimmune disorder that can be treated using a compound or a composition disclosed herein include an acute disseminated encephalomyelitis (ADEM), an Addison's disease, an allergy, allergic rhinitis, an Alzheimer's disease, an anti-phospholipid antibody syndrome (APS), an arthritis such as, e.g., a monoarthritis, an oligoarthritis, or a polyarthritis like an osteoarthritis, a rheumatoid arthritis, a juvenile idiopathic arthritis, a septic arthritis, a spondyloarthropathy, a gout, a pseudogout, or Still's disease, an asthma, an autoimmune deficiency syndrome (AIDS), an autoimmune hemolytic anemia, an autoimmune hepatitis, an autoimmune inner ear disease, a bullous pemphigoid, a celiac disease, a Chagas disease, a chronic obstructive pulmonary disease (COPD
  • a lupus nephritis a neonatal lupus, a subacute cutaneous lupus erythematosus, or a systemic lupus erythematosus, a morphea, a multiple sclerosis (MS), a myasthenia gravis, a myopathy such as, e.g., a dermatomyositis, an inclusion body myositis, or a polymyositis, a myositis, a narcolepsy, a neuromyotonia, a Parkinson's disease, a pemphigus vulgaris, a pernicious anaemia, a primary biliary cirrhosis, a psoriasis, a recurrent disseminated encephalomyelitis, a rheumatic fever, a schizophrenia, a scleroderma, a Sjögren's syndrome, a
  • Non-limiting examples of a symptom reduced by a method of treating an autoimmune disorder disclosed herein include inflammation, fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low or high blood pressure, irritability, anxiety, or depression, infertility or reduced sex drive (low libido), blood sugar changes, and depending on the type of autoimmune disease, an increase in the size of an organ or tissue, or the destruction of an organ or tissue.
  • Non-limiting examples of an inflammation symptom reduced by a method of treating an autoimmune disorder disclosed herein include edema, hyperemia, erythema, bruising, tenderness, stiffness, swollenness, fever, a chill, congestion of the respiratory tract including nose, and bronchi, congestion of a sinus, a breathing problem, fluid retention, a blood clot, a loss of appetite, an increased heart rate, a formation of granulomas, fibrinous, pus, or non-viscous serous fluid, a formation of an ulcer, or pain.
  • aspects of the present specification disclose a method of treating inflammation as a result of an autoimmune disorder, the method comprising the step of administering to an individual in need thereof a therapeutically effective amount of a RXR agonist, wherein administration of the compound or composition reduces a symptom associated with inflammation, thereby treating the individual.
  • aspects of the present specification also disclose a use of a RXR agonist to treat inflammation as a result of an autoimmune disorder, wherein administration of the compound or composition reduces a symptom associated with inflammation, thereby treating the individual.
  • Non-limiting examples of a RXR agonist include a compound or a composition disclosed herein.
  • Non-limiting examples of a symptom reduced by a method of treating inflammation disclosed herein include edema, hyperemia, erythema, bruising, tenderness, stiffness, swollenness, fever, a chill, congestion of the respiratory tract including nose, and bronchi, congestion of a sinus, a breathing problem, fluid retention, a blood clot, a loss of appetite, an increased heart rate, a formation of granulomas, fibrinous, pus, or non-viscous serous fluid, a formation of an ulcer, or pain.
  • Still aspects of the present specification disclose a method of treating a transplant rejection, the method comprising the step of administering to an individual in need thereof a therapeutically effective amount of a RXR agonist, wherein administration of the RXR agonist reduces a symptom associated with the transplant rejection, thereby treating the individual.
  • aspects of the present specification also disclose a use of a RXR agonist to treat a transplant rejection, wherein administration of the compound or composition reduces a symptom associated with the transplant rejection, thereby treating the individual.
  • Non-limiting examples of a RXR agonist include a compound or a composition disclosed herein.
  • Non-limiting examples of a transplant rejection include a hyperacute rejection, an acute rejection, or a chronic rejection, as well as, a graft-versus-host-disease.
  • Non-limiting examples of a symptom reduced by a method of treating a transplant rejection disclosed herein include inflammation, fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low or high blood pressure, irritability, anxiety, or depression, infertility or reduced sex drive (low libido), blood sugar changes, and depending on the type of autoimmune disease, an increase in the size of an organ or tissue, or the destruction of an organ or tissue.
  • Non-limiting examples of an inflammation symptom reduced by a method of treating a transplant rejection include edema, hyperemia, erythema, bruising, tenderness, stiffness, swollenness, fever, a chill, congestion of the respiratory tract including nose, and bronchi, congestion of a sinus, a breathing problem, fluid retention, a blood clot, a loss of appetite, an increased heart rate, a formation of granulomas, fibrinous, pus, or non-viscous serous fluid, a formation of an ulcer, or pain.
  • aspects of the present specification disclose a method of promoting Treg cell differentiation in an individual, the method comprising the step of administering to the individual in need thereof a therapeutically effective amount of a RXR agonist, wherein administration of the RXR agonist promotes Treg cell differentiation.
  • aspects of the present specification also disclose a use of a RXR agonist to promote Treg cell differentiation in an individual, wherein administration of the RXR agonist to the individual promotes Treg cell differentiation.
  • Administration of the RXR agonist to the individual can also inhibit Th17 cell differentiation.
  • aspects of the present specification disclose a method of inhibiting Th17 cell differentiation in an individual, the method comprising the step of administering to the individual in need thereof a therapeutically effective amount of a RXR agonist, wherein administration of the RXR agonist inhibits Th17 cell differentiation.
  • aspects of the present specification also disclose a use of a RXR agonist to inhibit Th17 cell differentiation in an individual, wherein administration of the RXR agonist to the individual inhibits Th17 cell differentiation.
  • Administration of the RXR agonist to the individual can also promote Treg cell differentiation.
  • aspects of the present specification disclose a method of concurrently promoting Treg cell differentiation as well as inhibiting Th17 cell differentiation in an individual, the method comprising the step of administering to the individual in need thereof a therapeutically effective amount of a RXR agonist, wherein administration of the RXR agonist promotes Treg cell differentiation and inhibits Th17 cell differentiation.
  • aspects of the present specification also disclose a use of a RXR agonist to concurrently promote Treg cell differentiation as well as inhibit Th17 cell differentiation in an individual, wherein administration of the RXR agonist to the individual promotes Treg cell differentiation and inhibits Th17 cell differentiation.
  • FIG. 1 shows RXR agonist effects on gene expression.
  • FIG. 1A shows that RXR agonists regulate Foxp3 expression; and
  • FIG. 1B shows that RXR agonists regulate ⁇ 4 ⁇ 7 (B) expression.
  • FIG. 2 shows RXR agonist effects on differentiation.
  • FIG. 2A shows that RXR agonists increase Treg differentiation under Th17 conditions; and
  • FIG. 2B shows that RXR agonists inhibit Th17 differentiation under Th17 conditions.
  • FIG. 3 shows the effects of RAR signaling inhibition on RXR agonist inducement of Treg differentiation.
  • FIG. 4 shows RXR agonist activation of transcription from RXR ⁇ , RXR ⁇ , RXR ⁇ , RAR ⁇ , RAR ⁇ , and RAR ⁇ using transactivation assays.
  • FIG. 5 shows that RXR agonists attenuate experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice.
  • FIG. 6 shows that RXR agonists reduce leukocyte infiltration into the central nervous system.
  • FIG. 6A shows that RXR agonists reduce CD4+ T cell infiltration into the central nervous system; and
  • FIG. 6B shows that RXR agonists reduce myeloid dendritic cell infiltration into the central nervous system.
  • FIG. 7 shows RXR agonists attenuate EAE in SJL mice.
  • RARs and RXRs and their cognate ligands function by distinct mechanisms.
  • the RARs always form heterodimers with RXRs and these RAR/RXR heterodimers bind to specific response elements in the promoter regions of target genes.
  • the binding of RAR agonists to the RAR receptor of the heterodimer results in activation of transcription of target genes leading to retinoid effects.
  • RXR agonists do not activate RAR/RXR heterodimers.
  • RXR heterodimer complexes like RAR/RXR can be referred to as non-permissive RXR heterodimers as activation of transcription due to ligand-binding occurs only at the non-RXR protein (e.g., RAR); activation of transcription due to ligand binding does not occur at the RXR.
  • RXRs also interact with nuclear receptors other than RARs and RXR agonists may elicit some of its biological effects by binding to such RXR/receptor complexes.
  • These RXR/receptor complexes can be referred to as permissive RXR heterodimers as activation of transcription due to ligand-binding could occur at the RXR, the other receptor, or both receptors.
  • RXR permissive heterodimers include, without limitation, peroxisome proliferator activated receptor/RXR(PPAR/RXR), farnesyl X receptor/RXR (FXR/RXR), or liver X receptor/RXR (LXR/RXR).
  • RXRs may form RXR/RXR homodimers which can be activated by RXR agonists leading to rexinoid effects.
  • RXRs interact with proteins other than nuclear receptors and ligand binding to an RXR within such protein complexes can also lead to rexinoid effects.
  • RXR agonists and RAR agonists elicit distinct biological outcomes and even in the instances where they mediate similar biological effects, they do so by different mechanisms.
  • the unwanted side effects of retinoids such as pro-inflammatory responses or mucocutaneous toxicity, are mediated by activation of one or more of the RAR receptor subtypes.
  • biological effects mediated via RXR pathways would not induce pro-inflammatory responses, and thus, would not result in unwanted side effects.
  • RXR agonists inhibit Th17 cell formation and promote Treg cell formation by mechanisms that do not involve their function as RAR agonists.
  • a selective RXR agonist that does not activate RARs would be a more effective agent in the treatment of an autoimmune disorder, inflammation associated with an autoimmune disorder, or a transplant rejection.
  • RXR agonists have cell differentiating effects in that they can regulate the Th17/Treg cell number ratio by elevating Treg cell numbers and suppressing Th17 cell numbers. In this manner, a normal balance of both these cell types can be achieved and immune homeostatis restored.
  • selective RXR agonists achieve these therapeutic effects without activation of RARs, they would be optimally effective and beneficial in treating an autoimmune disorder, inflammation associated with an autoimmune disorder, or a transplant rejection.
  • RXR agonist is synonymous with “RXR selective agonist” and refers to a compound that selectively binds to one or more RXR receptors like a RXR ⁇ , a RXR ⁇ , or a RXR ⁇ in a manner that elicits gene transcription via an RXR response element.
  • the term “selectively binds,” when made in reference to a RXR agonist, refers to the discriminatory binding of a RXR agonist to the indicated target receptor like a RXR ⁇ , a RXR ⁇ , or a RXR ⁇ such that the RXR agonist does not substantially bind with non-target receptors like a RAR ⁇ , a RAR ⁇ or a RAR ⁇ .
  • a RXR agonist may be a pure RXR agonist.
  • a pure RXR agonist is one which does not activate to any appreciable degree a permissive heterodimer such as, e.g., PPAR/RXR, FXR/RXR, and LXR/RXR.
  • PPAR/RXR PPAR/RXR
  • FXR/RXR FXR/RXR
  • LXR/RXR LXR/RXR.
  • One example of a pure RXR agonist is 3,7-dimethyl-6(S),7(S)-methano,7-[1,1,4,4-tetramethyl-1,2,3,4 tetrahydronaphth-7-yl]2(E),4(E) heptadienoic acid (RXR agonist 194204) disclosed herein, the structure of which is shown in Formula XXIX.
  • a pure RXR agonist shows no ability to activate a permissive heterodimer. In another aspect of this embodiment, a pure RXR agonist shows no ability to activate PPAR/RXR, FXR/RXR, and/or LXR/RXR. In other aspects of this embodiment, a pure RXR agonist activates a permissive heterodimer by 1% or less, 2% or less, 3% or less, 4% or less, 5% or less, 6% or less, 7% or less, 8% or less, 9% or less, or 10% or less relative to the ability of a non-pure RXR agonist to activate the same permissive heterodimer.
  • a non-pure RXR agonist is one that can activate a permissive heterodimer like PPAR/RXR, FXR/RXR, or LXR/RXR.
  • Example of a non-pure RXR agonist include, e.g., LGD1069 (bexarotene) and LGD268.
  • Binding affinity refers to the length of time a RXR agonist resides at its RXR receptor binding site, and can be viewed as the strength with which a RXR agonist binds its a RXR receptor. Binding affinity can be described as a RXR agonist's equilibrium dissociation constant (KD), which is defined as the ratio Kd/Ka at equilibrium, where Ka is a RXR agonist's association rate constant and kd is a RXR agonist's dissociation rate constant.
  • KD equilibrium dissociation constant
  • Binding affinity is determined by both the association and the dissociation and alone neither high association nor low dissociation can ensure high affinity.
  • the association rate constant (Ka), or on-rate constant (Kon) measures the number of binding events per unit time, or the propensity of a RXR agonist and its RXR receptor to associate reversibly into its agonist-receptor complex.
  • the association rate constant is expressed in M ⁇ 1 s ⁇ 1 , and is symbolized as follows: [Ag] ⁇ [Rc] ⁇ Kon. The larger the association rate constant, the more rapidly a RXR agonist binds to its RXR receptor, or the higher the binding affinity between agonist and receptor.
  • the dissociation rate constant measures the number of dissociation events per unit time propensity of an agonist-receptor complex to separate (dissociate) reversibly into its component molecules, namely the RXR agonist and the RXR receptor.
  • the dissociation rate constant is expressed in s ⁇ 1 , and is symbolized as follows: [Ag+Rc] ⁇ Koff. The smaller the dissociation rate constant, the more tightly bound a RXR agonist is to its RXR receptor, or the higher the binding affinity between agonsit and receptor.
  • the equilibrium dissociation constant measures the rate at which new agonist-receptor complexes formed equals the rate at which agonist-receptor complexes dissociate at equilibrium.
  • the smaller the equilibrium dissociation constant the more tightly bound a RXR agonist is to its RXR receptor, or the higher the binding affinity between agonist and receptor.
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an association rate constant of, e.g., less than 1 ⁇ 10 5 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 6 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 7 M ⁇ 1 s ⁇ 1 , or less than 1 ⁇ 10 8 M ⁇ 1 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an association rate constant of, e.g., more than 1 ⁇ 10 5 M ⁇ 1 s ⁇ 1 , more than 1 ⁇ 10 6 M ⁇ 1 s ⁇ 1 , more than 1 ⁇ 10 7 M ⁇ 1 s ⁇ 1 , or more than 1 ⁇ 10 8 M ⁇ 1 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an association rate constant between, e.g., 1 ⁇ 10 5 M ⁇ 1 s ⁇ 1 to 1 ⁇ 10 8 M ⁇ 1 s ⁇ 1 , 1 ⁇ 10 6 M ⁇ 1 s ⁇ 1 to 1 ⁇ 10 8 M ⁇ 1 s ⁇ 1 , 1 ⁇ 10 5 M ⁇ 1 s ⁇ 1 to 1 ⁇ 10 7 M ⁇ 1 s ⁇ 1 , or 1 ⁇ 10 6 M ⁇ 1 s ⁇ 1 to 1 ⁇ 10 7 M ⁇ 1 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have a disassociation rate constant of, e.g., less than 1 ⁇ 10 ⁇ 3 s ⁇ 1 , less than 1 ⁇ 10 ⁇ 4 s ⁇ 1 , or less than 1 ⁇ 10 ⁇ 5 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have a disassociation rate constant of, e.g., more than 1 ⁇ 10 ⁇ 3 s ⁇ 1 , more than 1 ⁇ 10 ⁇ 4 s ⁇ 1 , or more than 1 ⁇ 10 ⁇ 5 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have a disassociation rate constant between, e.g., 1 ⁇ 10 ⁇ 3 s ⁇ 1 to 1 ⁇ 10 ⁇ 5 s ⁇ 1 , 1 ⁇ 10 ⁇ 3 s ⁇ 1 to 1 ⁇ 10 ⁇ 4 s ⁇ 1 , or 1 ⁇ 10 ⁇ 4 s ⁇ 1 to 1 ⁇ 10 ⁇ 5 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an equilibrium disassociation constant of less than 100 nM. In aspects of this embodiment, the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an equilibrium disassociation constant of, e.g., less than 100 nM, less than 90 nM, less than 80 nM, less than 70 nM, less than 60 nM, less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, or less than 10 nM.
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an equilibrium disassociation between, e.g., 0.1 nM to 10 nM, 0.1 nM to 50 nM, 0.1 nM to 100 nM, 0.5 nM to 10 nM, 0.5 nM to 50 nM, 0.5 nM to 100 nM, 1 nM to 10 nM, 1 nM to 50 nM, or 1 nM to 100 nM.
  • the binding affinity of a RXR agonist that selectively binds to a RXR can have an association rate constant for a RAR receptor of, e.g., less than 1 ⁇ 10 0 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 1 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 2 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 3 M ⁇ 1 s ⁇ 1 , or less than 1 ⁇ 10 4 M ⁇ 1 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an association rate constant of a RAR receptor of, e.g., at most 1 ⁇ 10 0 M ⁇ 1 s ⁇ 1 , at most 1 ⁇ 10 1 M ⁇ 1 s ⁇ 1 , at most 1 ⁇ 10 2 M ⁇ 1 s ⁇ 1 , at most 1 ⁇ 10 3 M ⁇ 1 s ⁇ 1 , or at most 1 ⁇ 10 4 M ⁇ 1 s ⁇ 1 .
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an equilibrium disassociation constant for a RAR receptor of, e.g., more than 500 nM, for than 1,000 nM, more than 5,000 nm, or more than 10,000 nM.
  • the binding affinity of a RXR agonist that selectively binds to a RXR receptor can have an equilibrium disassociation constant for a RAR receptor between, e.g., 500 nM to 10,000 nM, 1,000 nM to 10,000 nM, or 5,000 nM to 10,000 nM.
  • Binding specificity is the ability of a RXR agonist to discriminate between a RXR receptor and a receptor that does not contain its binding site, such as, e.g., a RAR receptor.
  • One way to measure binding specificity is to compare the Kon association rate of a RXR agonist for its RXR relative to the Kon association rate of a RXR agonist for a receptor that does not contain its binding site. For example, comparing the association rate constant (Ka) of a RXR agonist for its RXR receptor relative to a RAR receptor
  • a RXR agonist that selectively binds to a RXR receptor can have an association rate constant (Ka) for a receptor not comprising its binding site of, e.g., less than 1 ⁇ 10 0 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 1 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 2 M ⁇ 1 s ⁇ 1 , less than 1 ⁇ 10 3 M ⁇ 1 s ⁇ 1 or less than 1 ⁇ 10 4 M ⁇ 1 s ⁇ 1 .
  • Ka association rate constant
  • a RXR agonist that selectively binds to a RXR receptor can have an association rate constant (Ka) for a receptor not comprising its binding site of, e.g., at most 1 ⁇ 10 0 M ⁇ 1 s ⁇ 1 , at most 1 ⁇ 10 1 M ⁇ 1 s ⁇ 1 , at most 1 ⁇ 10 2 M ⁇ 1 s ⁇ 1 , at most 1 ⁇ 10 3 M ⁇ 1 s ⁇ 1 or at most 1 ⁇ 10 4 M ⁇ 1 s ⁇ 1 .
  • Ka association rate constant
  • a RXR agonist that selectively binds to a RXR receptor can have an association rate constant (Ka) for a receptor not comprising its binding site of, e.g., at least 2-fold more, at least 3-fold more, at least 4-fold more, at least 5-fold more, at least 6-fold more, at least 7-fold more, at least 8-fold more, or at least 9-fold more.
  • a RXR agonist that selectively binds to a RXR receptor can have an association rate constant (Ka) for a receptor not comprising its binding site of, e.g., at least 10-fold more, at least 100-fold more, at least 1,000-fold more or at least 10,000-fold more.
  • a RXR agonist that selectively binds to a RXR receptor can have an association rate constant (Ka) for a receptor not comprising its binding site of, e.g., at most 1-fold more, at most 2-fold more, at most 3-fold more, at most 4-fold more, at most 5-fold more, at most 6-fold more, at most 7-fold more, at most 8-fold more, or at most 9-fold more.
  • Ka association rate constant
  • a RXR agonist that selectively binds to a RXR receptor can have an association rate constant (Ka) for a receptor not comprising its binding site of, e.g., at most 10-fold more, at most 100-fold more, at most 1,000-fold more or at most 10,000-fold more.
  • Ka association rate constant
  • the binding specificity of a RXR agonist that selectively binds to a RXR receptor can also be characterized as a binding ratio that such a RXR agonist can discriminate its RXR receptor relative to a receptor not comprising its binding site, such as, e.g., a RAR receptor.
  • a RXR agonist that selectively binds to a RXR receptor has a binding ratio for its RXR receptor relative to a receptor not comprising its binding site of, e.g., at least 2:1, at least 3:1, at least 4:1, at least 5:1, at least 64:1, at least 7:1, at least 8:1, at least 9:1, at least 10:1, at least 15:1, at least 20:1, at least 25:1, at least 30:1, at least 35:1, or at least 40:1.
  • a RXR agonist that selectively binds to a RXR receptor has a binding ratio for its RXR receptor relative to a RAR receptor of, e.g., at least 2:1, at least 3:1, at least 4:1, at least 5:1, at least 64:1, at least 7:1, at least 8:1, at least 9:1, at least 10:1, at least 15:1, at least 20:1, at least 25:1, at least 30:1, at least 35:1, or at least 40:1.
  • a RXR agonist will have a ratio of activity at a RXR receptor relative to a RAR receptor of, e.g., at least 5 greater, at least 10 greater, at least 15, or at least 20 greater.
  • the binding specificity of a RXR agonist that selectively binds to a RXR receptor can also be characterized as an activity ratio that such a RXR agonist can exert activity through binding to its RXR receptor relative to a receptor not comprising its binding site, such as, e.g., a RAR receptor.
  • a RXR agonist that selectively binds to a RXR receptor has an activity ratio through its RXR receptor relative to a receptor not comprising its binding site of, e.g., at least 2:1, at least 3:1, at least 4:1, at least 5:1, at least 64:1, at least 7:1, at least 8:1, at least 9:1, at least 10:1, at least 15:1, at least 20:1, at least 25:1, at least 30:1, at least 35:1, or at least 40:1.
  • a RXR agonist that selectively binds to a RXR receptor has an activity ratio through its RXR receptor relative to a RAR receptor of, e.g., at least 2:1, at least 3:1, at least 4:1, at least 5:1, at least 64:1, at least 7:1, at least 8:1, at least 9:1, at least 10:1, at least 15:1, at least 20:1, at least 25:1, at least 30:1, at least 35:1, or at least 40:1.
  • a RXR agonist is a compound having the structure of formula I:
  • Z is a radical having the structure of Formula II:
  • Y is cycloalkyl or cycloalkenyl of 3 to 8 carbons optionally substituted with one or two R 4 groups, or Y is selected from phenyl, pyridyl, thienyl, furyl, pyrrolyl, pyridazinyl, pyrimidiyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen, and
  • B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —
  • a RXR agonist is a compound having the structure of formula III:
  • R 2 is hydrogen or lower alkyl
  • R 3 is hydrogen or lower alkyl
  • B is hydrogen, COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or tri-lower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 8 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 8 is phenyl or lower alkylphenyl, R 9 and R 10 independently are hydrogen, an alkyl group of 1 to
  • a RXR agonist is a compound having the structure of formula IV:
  • n is 1 or 2; R 1 and R 2 independently are H, lower alkyl or fluoroalkyl; R 3 is hydrogen, lower alkyl, Cl or Br; R 4 is H, lower alkyl, fluoroalkyl or halogen, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 8 is an alkyl group of 1 to 10 carbons, or R 8 is phenyl or lower alkylphenyl, R 9 and R 10 independently are hydrogen, an alky
  • a RXR agonist is a compound having the structure of formula V:
  • R 4 is lower alkyl of 1 to 6 carbons
  • B is —COOH or —COOR 8 where R 8 is lower alkyl of 1 to 6 carbons, and the configuration about the cyclopropane ring is cis, and the configuration about the double bonds in the pentadienoic acid or ester chain attached to the cyclopropane ring is trans in each of the double bonds, or a pharmaceutically acceptable salt of the compound.
  • a RXR agonist is a compound having the structure of formula VI:
  • Z is a radical having the structure of Formula VII:
  • Y is cycloalkyl or cycloalkenyl of 3 to 8 carbons optionally substituted with one or two R 4 groups, or Y is selected from phenyl, pyridyl, thienyl, furyl, pyrrolyl, pyridazinyl, pyrimidiyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is S or O;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen
  • B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —
  • a RXR agonist is a compound having the structure of formula VIII:
  • X is S or O;
  • R 2 is hydrogen or lower alkyl;
  • R 3 is hydrogen or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, — COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 8 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 9 is phenyl or lower alkylphenyl, R 9 and R 10 independently
  • a RXR agonist is a compound having the structure of formula IX:
  • Z is a radical having the structure of Formula X:
  • Y is selected from pyridyl, pyrrolyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is NR 5 ;
  • n is 1 or 2;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen;
  • R 5 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11
  • a RXR agonist is a compound having the structure of formula IX:
  • Z is a radical having the structure of Formula X:
  • Y is selected from pyridyl, pyrrolyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is NR 5 ;
  • n is 1 or 2;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen;
  • R 5 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11
  • a RXR agonist is a compound having the structure of formula XII:
  • R is H, lower alkyl or 1 to 6 carbons, or a pharmaceutically acceptable salt of the compound.
  • a RXR agonist is a compound having the structure of formula XII:
  • Z is a radical having the structure of Formula XIV:
  • Y is cyclopropyl, the Y group being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is NR 5 ;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or hydrogen;
  • R 5 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O,
  • a RXR agonist is a compound having the structure of formula XV:
  • X is NR 5 ; R 5 is H or lower alkyl; R 2 is H or lower alkyl; R 3 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 9 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 9 is phenyl or lower alkylpheny
  • a RXR agonist is a compound having the structure of formula XVI:
  • Y is a bivalent radical having the structure of Formula XVII:
  • the two X 1 groups jointly represent an oxo ( ⁇ O) or thione ( ⁇ S) function, or X 1 is independently selected from H or alkyl of 1 to 6 carbons;
  • the two X 2 groups jointly represent an oxo ( ⁇ O) or a thione ( ⁇ S) function, or X 2 independently selected from H or alkyl of 1 to 6 carbons, with the proviso that one of the joint X 1 grouping or of the joint X 2 grouping represents an oxo ( ⁇ O) or thione ( ⁇ S) function;
  • W is O, C(R 1 ) 2 , or W does not exist;
  • R 1 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons;
  • R 2 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons;
  • R 3 is hydrogen, lower alkyl of 1 to 6 carbons, OR 1 , fluor
  • a RXR agonist is a compound having the structure of formula XVIII:
  • R 1 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons
  • R 1 * is hydrogen or C 1-6 -alkyl
  • R 2 * is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons
  • R 3 * is hydrogen, lower alkyl of 1 to 6 carbons, fluoro substituted lower alkyl of 1 to 6 carbons or halogen
  • X 1 * is an oxo ( ⁇ O) or a thione ( ⁇ S) group
  • A* is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , where R 8 is an alkyl group of 1 to 10 carbons or (trimethylsilyl)alkyl where the alkyl group has 1 to 10 carbons, or a cycloalkyl group of 5 to 10 carbons, or R 8 is phenyl or lower alkylphenyl
  • a RXR agonist is a compound having the structure of formulae XIX, XX, or XXI:
  • X is O, S, or (CR 1 R 1 ) n , where n is 0, 1 or 2;
  • Y is a bivalent radical having the structure of Formulae XXII or XXIII where o is an integer between 1 through 4
  • Y is a bivalent aryl or 5 or 6 membered heteroaryl radical having 1 to 3 heteroatoms selected from N, S and O, the aryl or heteroaryl groups being unsubstituted, or substituted with 1 to 3 C 1-6 alkyl or with 1 to 3 C 1-6 fluoroalkyl groups with the proviso that when the compound is in accordance with Formula II then Y is not a 5 or 6 membered ring;
  • X 1 is S or NH;
  • R 1 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons;
  • R 2 is independently H, lower alkyl of 1 to 6 carbons, OR 1 , adamantly, or lower fluoroalkyl of 1 to 6 carbons, or the two R 2 groups jointly represent an oxo ( ⁇ O) group with the proviso that when the compound is in accordance with Formula II then at least one of the R 2 substituents is branched-chain al
  • a RXR agonist is a compound having the structure of formula XXIV:
  • R is H, lower alkyl of 1 to 6 carbons, or a pharmaceutically acceptable salt of the compound.
  • a RXR agonist is a compound having the structure of formula XXV:
  • R is H, lower alkyl of 1 to 6 carbons, and R 1 is iso-propyl or tertiary-butyl, or a pharmaceutically acceptable salt of the compound.
  • a RXR agonist is a compound having the structure of formula XXVI:
  • R is H, lower alkyl of 1 to 6 carbons, and R 1 is iso-propyl, n-butyl or tertiary-butyl, or a pharmaceutically acceptable salt of the compound.
  • a RXR agonist is a compound having the structure of formula XXVII:
  • X is O or S
  • Y is a bivalent cycloalkyl or cycloalkenyl radical optionally substituted with one to four R 4 groups, the cycloalkenyl radical having 5 to 6 carbons and one double bond, or Y is a bivalent aryl or 5 or 6 membered heteroaryl radical having 1 to 3 heteroatoms selected from N, S and O, the aryl or heteroaryl groups optionally substituted with 1 to 4 R 4 groups with the proviso that the cycloalkyl or the cycloalkenyl radical is not substituted on the same carbon with the condensed cyclic moiety and with the diene containing moiety;
  • R 1 is independently H, alkyl of 1 to 6 carbons, or fluoroalkyl of 1 to 6 carbons;
  • R 2 is independently H, alkyl of 1 to 8 carbons, or fluoroalkyl of 1 to 8 carbons;
  • R 12 is independently H, alkyl of 1 to 8 carbons, or
  • R 4 is H, halogen, alkyl of 1 to 10 carbons, fluoro substituted alkyl of 1 to 6 carbons, alkoxy of 1 to 10 carbons, or alkylthio of 1 to 10 carbons; m is an integer having the values of 0 to 3; r is an integer having the values of 1 to 10; s is an integer having the values 1 to 4; t is an integer having the values 1 to 5;
  • R 9 represents a 5 or 6 membered heteroaryl ring having 1 to 3 heteroatoms selected from the group consisting of N, S and O;
  • B is hydrogen, COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 9 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 9 is phenyl or lower alkylphenyl, R 9
  • a RXR agonist is a compound having the structure of formula XXVIII:
  • R 1 is H or methyl
  • R 9 is H, alkyl of 1 to 6 carbons, or a pharmaceutically acceptable cation
  • R 3 is hydrogen, alkyl of 1 to 10 carbons, halogen, alkoxy of 1 to 10 carbons, or R 3 is selected from the groups shown below:
  • R 4 is H, halogen, alkyl of 1 to 10 carbons, carbons, alkoxy of 1 to 10; r is an integer having the values of 1 to 10; s is an integer having the values 1 to 4;
  • t represents a 5 or 6 membered heteroaryl ring having 1 to 3 heteroatoms selected from the group consisting of N, S and O, and t is an integer having the values 1 to 5.
  • a RXR agonist is 3,7-dimethyl-6(S),7(S)-methano,7-[1,1,4,4-tetramethyl-1,2,3,4-tetrahydronaphth-7-yl]2(E),4(E) heptadienoic acid, and has the structure of formula XXIX:
  • a RXR agonist having activity that promotes Treg cell differentiation.
  • a RXR agonist promotes Treg cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%.
  • a RXR agonist promotes Treg cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%.
  • a RXR agonist has activity that results in increased Foxp3 expression in cells exposed to the RXR agonist.
  • a RXR agonist increases Foxp3 expression in cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist increases Foxp3 expression in cells by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist has activity that results in increased Foxp3 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation conditions.
  • a RXR agonist increases Foxp3 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation not exposed to the same RXR agonist.
  • a RXR agonist increases Foxp3 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation not exposed to the same RXR agonist.
  • a RXR agonist has activity that results in increased ⁇ 4 ⁇ 7 expression in cells exposed to the RXR agonist.
  • a RXR agonist increases ⁇ 4 ⁇ 7 expression in cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist increases ⁇ 4 ⁇ 7 expression in cells by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist has activity that results in increased ⁇ 4 ⁇ 7 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation conditions.
  • a RXR agonist increases ⁇ 4 ⁇ 7 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation not exposed to the same RXR agonist.
  • a RXR agonist increases ⁇ 4 ⁇ 7 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation not exposed to the same RXR agonist.
  • a RXR agonist having activity that inhibits Th17 cell differentiation.
  • a RXR agonist inhibits Th17 cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%.
  • a RXR agonist inhibits Th17 cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%.
  • a RXR agonist has activity that results in decreased IL-17A expression in cells exposed to the RXR agonist.
  • a RXR agonist decreases IL-17A expression in cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist decreases IL-17A expression in cells by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist has activity that results in decreased IL-17A expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation conditions.
  • a RXR agonist decreases IL-17A expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation not exposed to the same RXR agonist.
  • a RXR agonist decreases IL-17A expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation not exposed to the same RXR agonist.
  • a RXR agonist having activity that both promotes Treg cell differentiation and inhibits Th17 cell differentiation.
  • a RXR agonist promotes Treg cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500% as well as inhibits Th17 cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%.
  • a RXR agonist promotes Treg cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, as well as inhibits Th17 cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 10%
  • a RXR agonist has activity that results in increased FoxP3 and/or ⁇ 4 ⁇ 7 expression as well as decreases IL-17A expression in cells exposed to the RXR agonist.
  • a RXR agonist increases FoxP3 and/or ⁇ 4 ⁇ 7 expression in cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, as well as decreases IL-17A expression in cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to cells not exposed to the same RXR agonist.
  • a RXR agonist increases FoxP3 and/or ⁇ 4 ⁇ 7 expression in cells by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, as well as decreases IL-17A expression in cells by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 20
  • a RXR agonist has activity that results in increased FoxP3 and/or ⁇ 4 ⁇ 7 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation conditions as well as decreases IL-17A expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation conditions.
  • a RXR agonist increases FoxP3 and/or ⁇ 4 ⁇ 7 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation not exposed to the same RXR agonist as well as decreases IL-17A expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Th17 cell differentiation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, at least 300%, at least 400%, or at least 500%, relative to naive CD4 + CD25
  • a RXR agonist increases FoxP3 and/or ⁇ 4 ⁇ 7 expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation by about 10% to about 25%, about 10% to about 50%, about 10% to about 75%, about 10% to about 100%, about 10% to about 200%, about 10% to about 300%, about 10% to about 400%, about 10% to about 500%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 200%, about 25% to about 300%, about 25% to about 400%, about 25% to about 500%, about 50% to about 100%, about 50% to about 200%, about 50% to about 300%, about 50% to about 400%, or about 50% to about 500%, relative to naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured under Treg cell differentiation not exposed to the same RXR agonist as well as decreases IL-17A expression in naive CD4 + CD25 ⁇ FoxP3 ⁇ cells cultured
  • compositions comprising a RXR agonist.
  • a RXR agonist includes the compounds disclosed herein.
  • the compositions disclosed herein may, or may not, comprise any number and combination of compounds disclosed herein.
  • a composition can comprise, e.g., two or more compounds disclosed herein, three or more compounds disclosed herein, four or more compounds disclosed herein, or five or more compounds disclosed herein.
  • a compound disclosed herein, or a composition comprising such a compound is generally administered to an individual as a pharmaceutical composition.
  • Pharmaceutical compositions may be prepared by combining a therapeutically effective amount of at least one compound as disclosed herein, or a pharmaceutically acceptable acid addition salt thereof, as an active ingredient, with conventional acceptable pharmaceutical excipients, and by preparation of unit dosage forms suitable for therapeutic use.
  • the term “pharmaceutical composition” and refers to a therapeutically effective concentration of an active compound, such as, e.g., any of the compounds disclosed herein.
  • the pharmaceutical composition does not produce an adverse, allergic, or other untoward or unwanted reaction when administered to an individual.
  • a pharmaceutical composition disclosed herein is useful for medical and veterinary applications.
  • a pharmaceutical composition may be administered to an individual alone, or in combination with other supplementary active compounds, agents, drugs or hormones.
  • the pharmaceutical compositions may be manufactured using any of a variety of processes, including, without limitation, conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, and lyophilizing.
  • the pharmaceutical composition can take any of a variety of forms including, without limitation, a sterile solution, suspension, emulsion, lyophilizate, tablet, pill, pellet, capsule, powder, syrup, elixir, or any other dosage form suitable for administration.
  • a pharmaceutical composition produced using the methods disclosed herein may be a liquid formulation, semi-solid formulation, or a solid formulation.
  • a formulation disclosed herein can be produced in a manner to form one phase, such as, e.g., an oil or a solid.
  • a formulation disclosed herein can be produced in a manner to form two phase, such as, e.g., an emulsion.
  • a pharmaceutical composition disclosed herein intended for such administration may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions.
  • Liquid formulations suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propylene glycol, polyethyleneglycol (PEG), glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • PEG polyethyleneglycol
  • glycerol glycerol
  • suitable mixtures thereof such as vegetable oils (such as olive oil)
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and
  • Semi-solid formulations suitable for topical administration include, without limitation, ointments, creams, salves, and gels.
  • the active compound may be admixed with at least one inert customary excipient (or carrier) such as, a lipid and/or polyethylene glycol.
  • Solid formulations suitable for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound may be admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate, (e) solution retarders, as for example, paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents,
  • a concentration of a therapeutic compound disclosed herein typically may be between about 50 mg/mL to about 1,000 mg/mL.
  • a therapeutically effective amount of a therapeutic compound disclosed herein may be from, e.g., about 50 mg/mL to about 100 mg/mL, about 50 mg/mL to about 200 mg/mL, about 50 mg/mL to about 300 mg/mL, about 50 mg/mL to about 400 mg/mL, about 50 mg/mL to about 500 mg/mL, about 50 mg/mL to about 600 mg/mL, about 50 mg/mL to about 700 mg/mL, about 50 mg/mL to about 800 mg/mL, about 50 mg/mL to about 900 mg/mL, about 50 mg/mL to about 1,000 mg/mL, about 100 mg/mL to about 200 mg/mL, about 100 mg/mL to about 300 mg/mL, about 100 mg/mL to about 400 mg/mL, about 100 mg/
  • an amount of a therapeutic compound disclosed herein typically may be between about 0.01% to about 45% by weight.
  • an amount of a therapeutic compound disclosed herein may be from, e.g., about 0.1% to about 45% by weight, about 0.1% to about 40% by weight, about 0.1% to about 35% by weight, about 0.1% to about 30% by weight, about 0.1% to about 25% by weight, about 0.1% to about 20% by weight, about 0.1% to about 15% by weight, about 0.1% to about 10% by weight, about 0.1% to about 5% by weight, about 1% to about 45% by weight, about 1% to about 40% by weight, about 1% to about 35% by weight, about 1% to about 30% by weight, about 1% to about 25% by weight, about 1% to about 20% by weight, about 1% to about 15% by weight, about 1% to about 10% by weight, about 1% to about 5% by weight, about 5% to about 45% by weight, about 5% to about 40% by weight, about 5% to about 35% by weight,
  • a pharmaceutical composition disclosed herein can optionally include a pharmaceutically acceptable carrier that facilitates processing of an active compound into pharmaceutically acceptable compositions.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio.
  • the term “pharmacologically acceptable carrier” is synonymous with “pharmacological carrier” and refers to any carrier that has substantially no long term or permanent detrimental effect when administered and encompasses terms such as “pharmacologically acceptable vehicle, stabilizer, diluent, additive, auxiliary, or excipient.”
  • a carrier generally is mixed with an active compound or permitted to dilute or enclose the active compound and can be a solid, semi-solid, or liquid agent. It is understood that the active compounds can be soluble or can be delivered as a suspension in the desired carrier or diluent.
  • aqueous media such as, e.g., water, saline, glycine, hyaluronic acid and the like
  • solid carriers such as, e.g., starch, magnesium stearate, mannitol, sodium saccharin, talcum, cellulose, glucose, sucrose, lactose, trehalose, magnesium carbonate, and the like
  • solvents dispersion media; coatings; antibacterial and antifungal agents; isotonic and absorption delaying agents; or any other inactive ingredient.
  • Selection of a pharmacologically acceptable carrier can depend on the mode of administration.
  • any pharmacologically acceptable carrier is incompatible with the active compound, its use in pharmaceutically acceptable compositions is contemplated.
  • Non-limiting examples of specific uses of such pharmaceutical carriers can be found in Pharmaceutical Dosage Forms and Drug Delivery Systems (Howard C. Ansel et al., eds., Lippincott Williams & Wilkins Publishers, 7 th ed. 1999); Remington: The Science and Practice of Pharmacy (Alfonso R. Gennaro ed., Lippincott, Williams & Wilkins, 20 th ed. 2000); Goodman & Gilman's The Pharmacological Basis of Therapeutics (Joel G.
  • a pharmaceutical composition disclosed herein can optionally include, without limitation, other pharmaceutically acceptable components (or pharmaceutical components), including, without limitation, buffers, preservatives, tonicity adjusters, salts, antioxidants, osmolality adjusting agents, physiological substances, pharmacological substances, bulking agents, emulsifying agents, wetting agents, sweetening or flavoring agents, and the like.
  • buffers include, without limitation, acetate buffers, borate buffers, citrate buffers, phosphate buffers, neutral buffered saline, and phosphate buffered saline.
  • antioxidants include, without limitation, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole, and butylated hydroxytoluene.
  • Useful preservatives include, without limitation, benzalkonium chloride, chlorobutanol, thimerosal, phenylmercuric acetate, phenylmercuric nitrate, a stabilized oxy chloro composition, such as, e.g., sodium chlorite and chelants, such as, e.g., DTPA or DTPA-bisamide, calcium DTPA, and CaNaDTPA-bisamide.
  • Tonicity adjustors useful in a pharmaceutical composition include, without limitation, salts such as, e.g., sodium chloride, potassium chloride, mannitol or glycerin and other pharmaceutically acceptable tonicity adjustor.
  • the pharmaceutical composition may be provided as a salt and can be formed with many acids, including but not limited to, hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms. It is understood that these and other substances known in the art of pharmacology can be included in a pharmaceutical composition useful in the invention.
  • a compound disclosed herein, or a composition comprising such a compound may also be incorporated into a drug delivery platform in order to achieve a controlled compound release profile over time.
  • a drug delivery platform comprises a compound disclosed herein dispersed within a polymer matrix, typically a biodegradable, bioerodible, and/or bioresorbable polymer matrix.
  • polymer refers to synthetic homo- or copolymers, naturally occurring homo- or copolymers, as well as synthetic modifications or derivatives thereof having a linear, branched or star structure. Copolymers can be arranged in any form, such as, e.g., random, block, segmented, tapered blocks, graft, or triblock. Polymers are generally condensation polymers. Polymers can be further modified to enhance their mechanical or degradation properties by introducing cross-linking agents or changing the hydrophobicity of the side residues. If crosslinked, polymers are usually less than 5% crosslinked, usually less than 1% crosslinked.
  • Suitable polymers include, without limitation, alginates, aliphatic polyesters, polyalkylene oxalates, polyamides, polyamidoesters, polyanhydrides, polycarbonates, polyesters, polyethylene glycol, polyhydroxyaliphatic carboxylic acids, polyorthoesters, polyoxaesters, polypeptides, polyphosphazenes, polysaccharides, and polyurethanes.
  • the polymer usually comprises at least about 10% (w/w), at least about 20% (w/w), at least about 30% (w/w), at least about 40% (w/w), at least about 50% (w/w), at least about 60% (w/w), at least about 70% (w/w), at least about 80% (w/w), or at least about 90% (w/w) of the drug delivery platform.
  • biodegradable, bioerodible, and/or bioresorbable polymers and methods useful to make a drug delivery platform are described in, e.g., Drost, et. al., Controlled Release Formulation, U.S. Pat. No. 4,756,911; Smith, et. al., Sustained Release Drug Delivery Devices, U.S. Pat. No.
  • a polymer composing the matrix is a polypeptide such as, e.g., silk fibroin, keratin, or collagen.
  • a polymer composing the matrix is a polysaccharide such as, e.g., cellulose, agarose, elastin, chitosan, chitin, or a glycosaminoglycan like chondroitin sulfate, dermatan sulfate, keratan sulfate, or hyaluronic acid.
  • a polymer composing the matrix is a polyester such as, e.g., D-lactic acid, L-lactic acid, racemic lactic acid, glycolic acid, caprolactone, and combinations thereof.
  • a suitable polymer for forming a suitable disclosed drug delivery platform depends on several factors.
  • the more relevant factors in the selection of the appropriate polymer(s) include, without limitation, compatibility of polymer with drug, desired release kinetics of drug, desired biodegradation kinetics of platform at implantation site, desired bioerodible kinetics of platform at implantation site, desired bioresorbable kinetics of platform at implantation site, in vivo mechanical performance of platform, processing temperatures, biocompatibility of platform, and patient tolerance.
  • Other relevant factors that, to some extent, dictate the in vitro and in vivo behavior of the polymer include the chemical composition, spatial distribution of the constituents, the molecular weight of the polymer and the degree of crystallinity.
  • a drug delivery platform includes both a sustained release drug delivery platform and an extended release drug delivery platform.
  • sustained release refers to the release of a compound disclosed herein over a period of about seven days or more.
  • extended release refers to the release of a compound disclosed herein over a period of time of less than about seven days.
  • a sustained release drug delivery platform releases a compound disclosed herein with substantially first order release kinetics over a period of, e.g., about 7 days after administration, about 15 days after administration, about 30 days after administration, about 45 days after administration, about 60 days after administration, about 75 days after administration, or about 90 days after administration.
  • a sustained release drug delivery platform releases a compound disclosed herein with substantially first order release kinetics over a period of, e.g., at least 7 days after administration, at least 15 days after administration, at least 30 days after administration, at least 45 days after administration, at least 60 days after administration, at least 75 days after administration, or at least 90 days after administration.
  • a drug delivery platform releases a compound disclosed herein with substantially first order release kinetics over a period of, e.g., about 1 day after administration, about 2 days after administration, about 3 days after administration, about 4 days after administration, about 5 days after administration, or about 6 days after administration.
  • a drug delivery platform releases a compound disclosed herein with substantially first order release kinetics over a period of, e.g., at most 1 day after administration, at most 2 days after administration, at most 3 days after administration, at most 4 days after administration, at most 5 days after administration, or at most 6 days after administration.
  • an autoimmune disorder arises from an overactive immune response of the body against substances and tissues normally present in the body resulting in a break in tolerance toward self-antigens. In other words, the body actually attacks its own cells because the immune system mistakes some part of the body as a pathogen and attacks it. Characterized by the development of pathogenic T cell populations infiltrating the target organ or tissue, autoimmune disorders may be restricted to certain organs or involve a particular tissue in different places.
  • Autoimmune diseases can be broadly divided into systemic and organ-specific autoimmune disorders, depending on the principal clinico-pathologic features of each disease.
  • Systemic autoimmune diseases include, without limitation, systemic lupus erythematosus (SLE), Sjögren's syndrome, Scleroderma, rheumatoid arthritis and polymyositis.
  • Local autoimmune diseases may be endocrinologic (Diabetes Mellitus Type 1, Hashimoto's thyroiditis, Addison's disease etc.), dermatologic (pemphigus vulgaris), hematologic (autoimmune haemolytic anemia), neural (multiple sclerosis) or can involve virtually any circumscribed mass of body tissue.
  • Non-limiting examples of an autoimmune disorder that can be treated using a compound or a composition disclosed herein include an acute disseminated encephalomyelitis (ADEM), an Addison's disease, an allergy, allergic rhinitis, an Alzheimer's disease, an anti-phospholipid antibody syndrome (APS), an arthritis such as, e.g., a monoarthritis, an oligoarthritis, or a polyarthritis like an osteoarthritis, a rheumatoid arthritis, a juvenile idiopathic arthritis, a septic arthritis, a spondyloarthropathy, a gout, a pseudogout, or Still's disease, an asthma, an autoimmune deficiency syndrome (AIDS), an autoimmune hemolytic anemia, an autoimmune hepatitis, an autoimmune inner ear disease, a bullous pemphigoid, a celiac disease, a Chagas disease, a chronic obstructive pulmonary disease (COPD
  • a lupus nephritis a neonatal lupus, a subacute cutaneous lupus erythematosus, or a systemic lupus erythematosus, a morphea, a multiple sclerosis (MS), a myasthenia gravis, a myopathy such as, e.g., a dermatomyositis, an inclusion body myositis, or a polymyositis, a myositis, a narcolepsy, a neuromyotonia, a Parkinson's disease, a pemphigus vulgaris, a pernicious anaemia, a primary biliary cirrhosis, a psoriasis, a recurrent disseminated encephalomyelitis, a rheumatic fever, a schizophrenia, a scleroderma, a Sjögren's syndrome, a
  • Arthritis includes a group of conditions involving damage to the joints of the body due to the inflammation of the synovium including, without limitation osteoarthritis, rheumatoid arthritis, juvenile idiopathic arthritis, spondyloarthropathies like ankylosing spondylitis, reactive arthritis (Reiter's syndrome), psoriatic arthritis, enteropathic arthritis associated with inflammatory bowel disease, Whipple's disease and Behcet's disease, septic arthritis, gout (also known as gouty arthritis, crystal synovitis, metabolic arthritis), pseudogout (calcium pyrophosphate deposition disease), and Still's disease. Arthritis can affect a single joint (monoarthritis), two to four joints (oligoarthritis) or five or more joints (polyarthritis) and can be either an auto-immune disease or a non-autoimmune disease.
  • Myopathies are caused by problems with the immune system attacking components of the muscle, leading to signs of inflammation in the muscle Inflammatory myopathies include, without limitation, dermatomyositis, inclusion body myositis, and polymyositis.
  • Vasculitis is a varied group of disorders featuring inflammation of a vessel wall including lymphatic vessels and blood vessels like veins (phlebitis), arteries (arteritis) and capillaries due to leukocyte migration and resultant damage.
  • the inflammation may affect any size blood vessel, anywhere in the body. It may affect either arteries and/or veins.
  • the inflammation may be focal, meaning that it affects a single location within a vessel; or it may be widespread, with areas of inflammation scattered throughout a particular organ or tissue, or even affecting more than one organ system in the body.
  • Vasculitis include, without limitation, Buerger's disease (thromboangiitis obliterans), cerebral vasculitis (central nervous system vasculitis), Churg-Strauss arteritis, cryoglobulinemia, essential cryoglobulinemic vasculitis, giant cell (temporal) arteritis, Golfers vasculitis, Henoch-Schonlein purpura, hypersensitivity vasculitis (allergic vasculitis), Kawasaki disease, microscopic polyarteritis/polyangiitis, polyarteritis nodosa, polymyalgia rheumatica (PMR), rheumatoid vasculitis, Takayasu arteritis, Wegener's granulomatosis, and vasculitis secondary to connective tissue disorders like systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), relapsing polychondritis, Behçet
  • autoimmune disorder is a skin disorder.
  • Skin disorders include, without limitation, a dermatitis, including chronic actinic dermatitis, an eczema like atopic eczema, contact eczema, xerotic eczema, seborrhoeic dermatitis, dyshidrosis, discoid eczema, venous eczema, dermatitis herpetiformis, neurodermatitis, and autoeczematization, and statis dermatitis, hidradenitis suppurativa, psoriasis including plaqure psoriasis, nail psoriasis, guttate psoriasis, scalp psoriasis, inverse psoriasis, pustular psoriasis, and erythrodermis psoriasis, rosacea and scleroderma including morphea.
  • a gastrointestinal disorder includes, without limitation, irritable bowel disease, an inflammatory bowel disease including Crohn's disease and an ulcerative colitis like ulcerative proctitis, left-sided colitis, pancolitis and fulminant colitis.
  • Transplant rejection occurs when a transplanted organ or tissue is not accepted by the body of the transplant recipient because the immune system of the recipient attacks the transplanted organ or tissue.
  • An adaptive immune response, transplant rejection is mediated through both T cell mediated and humoral immune (antibodies) mechanisms.
  • the number of mismatched alleles determines the speed and magnitude of the rejection response. Different mechanisms tend to act against different transplants.
  • a transplant rejection can be classified as a hyperacute rejection, an acute rejection, or a chronic rejection.
  • Hyperacute rejection is a complement-mediated response in recipients with pre-existing antibodies to the donor (for example, ABO blood type antibodies). Hyperacute rejection occurs within minutes after the transplant and must be immediately removed to prevent a severe systemic inflammatory response. Rapid agglutination of the blood occurs.
  • Acute rejection may begin as early as one week after transplantation (as opposed to hyperacute rejection, which is immediate).
  • the risk of acute rejection is highest in the first three months after transplantation.
  • acute rejection can also occur months to years after transplantation.
  • the reason that acute rejection usually begins one week after transplantation is that T-cells are involved in the rejection mechanism. These T-cells must differentiate before rejection begins.
  • the T-cells cause cells in the transplanted tissue to lyse, or produce cytokines that cause necrosis of the transplanted tissue.
  • a single episode of acute rejection is not a cause for concern if recognized and treated promptly, and rarely leads to organ failure.
  • Acute rejection occurs to some degree in all transplants (except those between identical twins) unless the immune response in altered through the use of immunosuppressive drugs. It is caused by mismatched HLA, which are present on all cells of the body. There are a large number of different alleles of each HLA, so a perfect match between all HLA in the donor tissue and the recipient's body is extremely rare.
  • Chronic rejection of a transplanted organ or tissue is where the rejection is due to a poorly understood chronic inflammatory and immune response against the transplanted tissue.
  • Chronic rejection after lung transplantation is the leading cause of long-term morbidity and mortality in lung transplant patients
  • GVHD graft-versus-host disease
  • aGVHD acute or fulminant form of the disease
  • cGVHD chronic graft-versus-host-disease
  • Acute GVHD is characterized by selective damage to the liver, skin and mucosa, gastrointestinal tract, immune system (the hematopoietic system, e.g., the bone marrow and the thymus) itself, and the lungs in the form of idiopathic pneumonitis.
  • Acute GVHD of the GI tract can result in severe intestinal inflammation, sloughing of the mucosal membrane, severe diarrhea, abdominal pain, nausea, and vomiting. This is typically diagnosed via intestinal biopsy.
  • Liver GVHD is measured by the bilirubin level in acute patients. Skin GVHD results in a diffuse maculopapular rash, sometimes in a lacy pattern.
  • Acute GVHD is staged as follows: overall grade (skin-liver-gut) with each organ staged individually from a low of 1 to a high of 4. Patients with grade IV GVHD usually have a poor prognosis. If the GVHD is severe and requires intense immunosuppression involving steroids and additional agents to get under control, the patient may develop severe infections as a result of the immunosuppression and may die of infection. Chronic GVHD also attacks the above organs, but over its long-term course can also cause damage to the connective tissue and exocrine glands.
  • aspects of the present invention provide, in part, reducing a symptom associated with an autoimmune disorder or transplant rejection.
  • the actual symptoms associated with an autoimmune disorder or transplant rejection disclosed herein are well known and can be determined by a person of ordinary skill in the art by taking into account factors, including, without limitation, the location of the autoimmune disorder or transplant rejection, the cause of the autoimmune disorder or transplant rejection, the severity of the autoimmune disorder or transplant rejection, the tissue or organ affected by the autoimmune disorder or transplant rejection, and the autoimmune disorder or transplant rejection associated with the inflammation.
  • Non-limiting examples of a symptom reduced by a method of treating an autoimmune disorder or transplant rejection disclosed herein include inflammation, fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low or high blood pressure, irritability, anxiety, or depression, infertility or reduced sex drive (low libido), blood sugar changes, and depending on the type of autoimmune disorder or transplant rejection, an increase in the size of an organ or tissue, or the destruction of an organ or tissue.
  • Non-limiting examples of an inflammation symptom reduced by a method of treating an autoimmune disorder disclosed herein include edema, hyperemia, erythema, bruising, tenderness, stiffness, swollenness, fever, a chill, congestion of the respiratory tract including nose and/or bronchi, congestion of a sinus, a breathing problem, fluid retention, a blood clot, a loss of appetite, an increased heart rate, a formation of granulomas, fibrinous, pus, or non-viscous serous fluid, a formation of an ulcer, or pain.
  • aspects of the present invention provide, in part, a mammal.
  • a mammal includes a human, and a human can be a patient.
  • Other aspects of the present invention provide, in part, an individual.
  • An individual includes a mammal and a human, and a human can be a patient.
  • administering means any delivery mechanism that provides a compound or a composition disclosed herein to an individual that potentially results in a clinically, therapeutically, or experimentally beneficial result.
  • Administration of a compound or a composition disclosed herein include a variety of enteral or parenteral approaches including, without limitation, oral administration in any acceptable form, such as, e.g., tablet, liquid, capsule, powder, or the like; topical administration in any acceptable form, such as, e.g., drops, spray, creams, gels or ointments; buccal, nasal, and/or inhalation administration in any acceptable form; rectal administration in any acceptable form; vaginal administration in any acceptable form; intravascular administration in any acceptable form, such as, e.g., intravenous bolus injection, intravenous infusion, intra-arterial bolus injection, intra-arterial infusion and catheter instillation into the vasculature; peri- and intra-tissue administration in any acceptable form, such as, e.g., intraperitoneal injection, intramuscular injection, subcutaneous injection, subcutaneous infusion, intraocular injection, retinal injection, or sub-retinal injection or epidural injection; intravesicular
  • biodegradable polymers and methods of use are described in, e.g., Handbook of Biodegradable Polymers (Abraham J. Domb et al., eds., Overseas Publishers Association, 1997).
  • a compound or a composition disclosed herein can be administered to a mammal using a variety of routes.
  • Routes of administration suitable for treating an autoimmune disorder or transplant rejection as disclosed herein include both local and systemic administration. Local administration results in significantly more delivery of a composition to a specific location as compared to the entire body of the mammal, whereas, systemic administration results in delivery of a composition to essentially the entire body of the individual.
  • Routes of administration suitable for or treating an autoimmune disorder or transplant rejection as disclosed herein also include both central and peripheral administration. Central administration results in delivery of a compound or a composition to essentially the central nervous system of the individual and includes, e.g., intrathecal administration, epidural administration as well as a cranial injection or implant.
  • Peripheral administration results in delivery of a compound or a composition to essentially any area of an individual outside of the central nervous system and encompasses any route of administration other than direct administration to the spine or brain.
  • the actual route of administration of a compound or a composition disclosed herein used can be determined by a person of ordinary skill in the art by taking into account factors, including, without limitation, the type of an autoimmune disorder or transplant rejection, the location of the autoimmune disorder or transplant rejection, the cause of the autoimmune disorder or transplant rejection, the severity of the autoimmune disorder or transplant rejection, the duration of treatment desired, the degree of relief desired, the duration of relief desired, the particular compound or composition used, the rate of excretion of the compound or composition used, the pharmacodynamics of the compound or composition used, the nature of the other compounds to be included in the composition, the particular route of administration, the particular characteristics, history and risk factors of the individual, such as, e.g., age, weight, general health and the like, the response of the individual to the treatment, or any combination thereof.
  • a compound or a composition disclosed herein is administered systemically to a mammal. In another embodiment, a compound or a composition disclosed herein is administered locally to a mammal. In an aspect of this embodiment, a compound or a composition disclosed herein is administered to a site of autoimmune disorder or transplant rejection of a mammal. In another aspect of this embodiment, a compound or a composition disclosed herein is administered to the area surrounding an autoimmune disorder or transplant rejection of a mammal.
  • aspects of the present specification provide, in part, administering a therapeutically effective amount of a compound or a composition disclosed herein.
  • therapeutically effective amount is synonymous with “therapeutically effective dose” and when used in reference to treating an autoimmune disorder means the minimum dose of a compound or composition disclosed herein necessary to achieve the desired therapeutic effect and includes a dose sufficient to reduce a symptom associated with an autoimmune disorder or transplant rejection.
  • a therapeutically effective amount of a compound or a composition disclosed herein reduces a symptom associated with an autoimmune disorder or transplant rejection by, e.g., at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or at least 100%.
  • a therapeutically effective amount of a compound or a composition disclosed herein reduces a symptom associated with an autoimmune disorder or transplant rejection by, e.g., at most 10%, at most 20%, at most 30%, at most 40%, at most 50%, at most 60%, at most 70%, at most 80%, at most 90% or at most 100%.
  • a therapeutically effective amount of a compound or a composition disclosed herein reduces a symptom associated with an autoimmune disorder or transplant rejection by, e.g., about 10% to about 100%, about 10% to about 90%, about 10% to about 80%, about 10% to about 70%, about 10% to about 60%, about 10% to about 50%, about 10% to about 40%, about 20% to about 100%, about 20% to about 90%, about 20% to about 80%, about 20% to about 20%, about 20% to about 60%, about 20% to about 50%, about 20% to about 40%, about 30% to about 100%, about 30% to about 90%, about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, or about 30% to about 50%.
  • a therapeutically effective amount of a compound or a composition disclosed herein is the dosage sufficient to reduces a symptom associated with an autoimmune disorder or transplant rejection for, e.g., at least one week, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, at least seven months, at least eight months, at least nine months, at least ten months, at least eleven months, or at least twelve months.
  • the amount of active component in a compound or a composition disclosed herein for treating an autoimmune disorder or transplant rejection can be varied so that a suitable dosage is obtained.
  • the actual therapeutically effective amount of a compound or a composition disclosed herein to be administered to a mammal can be determined by a person of ordinary skill in the art by taking into account factors, including, without limitation, the type of the autoimmune disorder or transplant rejection, the location of the autoimmune disorder or transplant rejection, the cause of the autoimmune disorder or transplant rejection, the severity of the autoimmune disorder or transplant rejection, the duration of treatment desired, the degree of relief desired, the duration of relief desired, the particular compound or composition used, the rate of excretion of the compound or composition used, the pharmacodynamics of the compound or composition used, the nature of the other compounds to be included in the composition, the particular route of administration, the particular characteristics, history and risk factors of the individual, such as, e.g., age, weight, general health and the like, the response of the individual to the treatment, or any combination thereof.
  • the actual effect amount of a compound or a composition disclosed herein will further depend upon factors, including, without limitation, the frequency of administration, the half-life of the compound or composition disclosed herein, or any combination thereof.
  • an effective amount of a compound or a composition disclosed herein can be extrapolated from in vitro assays and in vivo administration studies using animal models prior to administration to humans. Wide variations in the necessary effective amount are to be expected in view of the differing efficiencies of the various routes of administration. For instance, oral administration generally would be expected to require higher dosage levels than administration by intravenous or intravitreal injection. Variations in these dosage levels can be adjusted using standard empirical routines of optimization, which are well-known to a person of ordinary skill in the art. The precise therapeutically effective dosage levels and patterns are preferably determined by the attending physician in consideration of the above-identified factors.
  • a therapeutically effective amount when administering a compound or a composition disclosed herein to a mammal, a therapeutically effective amount generally is in the range of about 0.001 mg/kg/day to about 100.0 mg/kg/day.
  • an effective amount of a compound or a composition disclosed herein can be, e.g., about 0.01 mg/kg/day to about 0.1 mg/kg/day, about 0.03 mg/kg/day to about 3.0 mg/kg/day, about 0.1 mg/kg/day to about 3.0 mg/kg/day, or about 0.3 mg/kg/day to about 3.0 mg/kg/day.
  • a therapeutically effective amount of a compound or a composition disclosed herein can be, e.g., at least 0.001 mg/kg/day, at least 0.01 mg/kg/day, at least 0.1 mg/kg/day, at least 1.0 mg/kg/day, at least 10 mg/kg/day, or at least 100 mg/kg/day.
  • a therapeutically effective amount of a compound or a composition disclosed herein can be, e.g., at most 0.001 mg/kg/day, at most 0.01 mg/kg/day, at most 0.1 mg/kg/day, at most 1.0 mg/kg/day, at most 10 mg/kg/day, or at most 100 mg/kg/day.
  • a therapeutically effective amount when administering a compound or a composition disclosed herein to a mammal, a therapeutically effective amount generally is in the range of about 0.001 mg/m 2 /day to about 100.0 mg/m 2 /day.
  • an effective amount of a compound or a composition disclosed herein can be, e.g., about 0.01 mg/m 2 /day to about 0.1 mg/m 2 /day, about 0.03 mg/m 2 /day to about 3.0 mg/m 2 /day, about 0.1 mg/m 2 /day to about 3.0 mg/m 2 /day, or about 0.3 mg/m 2 /day to about 3.0 mg/m 2 /day.
  • a therapeutically effective amount of a compound or a composition disclosed herein can be, e.g., at least 0.001 mg/m 2 /day, at least 0.01 mg/m 2 /day, at least 0.1 mg/m 2 /day, at least 1.0 mg/m 2 /day, at least 10 mg/m 2 /day, or at least 100 mg/m 2 /day.
  • a therapeutically effective amount of a compound or a composition disclosed herein can be, e.g., at most 0.001 mg/m 2 /day, at most 0.01 mg/m 2 /day, at most 0.1 mg/m 2 /day, at most 1.0 mg/m 2 /day, at most 10 mg/m 2 /day, or at most 100 mg/m 2 /day.
  • Dosing can be single dosage or cumulative (serial dosing), and can be readily determined by one skilled in the art.
  • treatment of an autoimmune disorder or transplant rejection may comprise a one-time administration of an effective dose of a compound or a composition disclosed herein.
  • an effective dose of a compound or a composition disclosed herein can be administered once to a mammal, e.g., as a single injection or deposition at or near the site exhibiting a symptom of an autoimmune disorder or transplant rejection or a single oral administration of the compound or a composition.
  • treatment of an autoimmune disorder or transplant rejection may comprise multiple administrations of an effective dose of a compound or a composition disclosed herein carried out over a range of time periods, such as, e.g., daily, once every few days, weekly, monthly or yearly.
  • a compound or a composition disclosed herein can be administered once or twice weekly to a mammal.
  • the timing of administration can vary from mammal to mammal, depending upon such factors as the severity of a mammal's symptoms.
  • an effective dose of a compound or a composition disclosed herein can be administered to a mammal once a month for an indefinite period of time, or until the mammal no longer requires therapy.
  • a person of ordinary skill in the art will recognize that the condition of the mammal can be monitored throughout the course of treatment and that the effective amount of a compound or a composition disclosed herein that is administered can be adjusted accordingly.
  • a compound or a composition disclosed herein as disclosed herein can also be administered to a mammal in combination with other therapeutic compounds to increase the overall therapeutic effect of the treatment.
  • the use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
  • Y is cycloalkyl or cycloalkenyl of 3 to 8 carbons optionally substituted with one or two R 4 groups, or Y is selected from phenyl, pyridyl, thienyl, furyl, pyrrolyl, pyridazinyl, pyrimidiyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen, and
  • B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —
  • Y is selected from thienyl and furyl, the groups being optionally with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen, and
  • B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —OCOR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or tri-lower alkylsilyl, where R 7 is an al
  • R 2 is hydrogen or lower alkyl
  • R 3 is hydrogen or lower alkyl
  • B is hydrogen, COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or tri-lower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 9 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 9 is phenyl or lower alkylphenyl, R 9 and R 10 independently are hydrogen, an alkyl group of 1 to
  • n is 1 or 2; R 1 and R 2 independently are H, lower alkyl or fluoroalkyl; R 3 is hydrogen, lower alkyl, Cl or Br; R 4 is H, lower alkyl, fluoroalkyl or halogen, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 9 is an alkyl group of 1 to 10 carbons, or R 8 is phenyl or lower alkylphenyl, R 9 and R 10 independently are hydrogen, an alky
  • R 4 is lower alkyl of 1 to 6 carbons
  • B is —COOH or —COOR 8 where R 8 is lower alkyl of 1 to 6 carbons, and the configuration about the cyclopropane ring is cis, and the configuration about the double bonds in the pentadienoic acid or ester chain attached to the cyclopropane ring is trans in each of the double bonds, or a pharmaceutically acceptable salt of the compound.
  • Y is cycloalkyl or cycloalkenyl of 3 to 8 carbons optionally substituted with one or two R 4 groups, or Y is selected from phenyl, pyridyl, thienyl, furyl, pyrrolyl, pyridazinyl, pyrimidiyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is S or O;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen
  • B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —
  • X is S or O;
  • R 2 is hydrogen or lower alkyl;
  • R 3 is hydrogen or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, — COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 9 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 9 is phenyl or lower alkylphenyl, R 9 and R 10 independently
  • Y is selected from pyridyl, pyrrolyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is NR 5 ;
  • n is 1 or 2;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen;
  • R 5 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11
  • Y is selected from pyridyl, pyrrolyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, and imidazolyl, the groups being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is NR 5 ;
  • n is 1 or 2;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalkyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or halogen;
  • R 5 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11
  • R is H, lower alkyl or 1 to 6 carbons, or a pharmaceutically acceptable salt of the compound.
  • Y is cyclopropyl, the Y group being optionally substituted with one or two R 4 groups, the divalent Y radical being substituted by the Z and —(CR 1 ⁇ CR 1 ⁇ CR 1 ⁇ CR 1 )— groups on adjacent carbons;
  • X is NR 5 ;
  • R 1 and R 2 independently are H, lower alkyl or fluoroalyl;
  • R 3 is hydrogen, lower alkyl, Cl or Br;
  • R 4 is lower alkyl, fluoroalkyl or hydrogen;
  • R 5 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O,
  • X is NR 5 ; R 5 is H or lower alkyl; R 2 is H or lower alkyl; R 3 is H or lower alkyl, and B is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , —CH 2 OH, —CH 2 OR 11 , —CH 2 OCOR 11 , —CHO, —CH(OR 12 ) 2 , —CHOR 13 O, —COR 7 , —CR 7 (OR 12 ) 2 , —CR 7 OR 13 O, or trilower alkylsilyl, where R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R 8 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 8 is phenyl or lower alkylpheny
  • the two X 1 groups jointly represent an oxo ( ⁇ O) or thione ( ⁇ S) function, or X 1 is independently selected from H or alkyl of 1 to 6 carbons;
  • the two X 2 groups jointly represent an oxo ( ⁇ O) or a thione ( ⁇ S) function, or X 2 independently selected from H or alkyl of 1 to 6 carbons, with the proviso that one of the joint X 1 grouping or of the joint X 2 grouping represents an oxo ( ⁇ O) or thione ( ⁇ S) function;
  • W is O, C(R 1 ) 2 , or W does not exist;
  • R 1 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons;
  • R 2 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons;
  • R 3 is hydrogen, lower alkyl of 1 to 6 carbons, OR 1 , fluor
  • R 1 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons
  • R 1 * is hydrogen or C 1-6 -alkyl
  • R 2 * is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons
  • R 3 * is hydrogen, lower alkyl of 1 to 6 carbons, fluoro substituted lower alkyl of 1 to 6 carbons or halogen
  • X 1 * is an oxo ( ⁇ O) or a thione ( ⁇ S) group
  • A* is hydrogen, —COOH or a pharmaceutically acceptable salt thereof, —COOR 8 , —CONR 9 R 10 , where R 8 is an alkyl group of 1 to 10 carbons or (trimethylsilyl)alkyl where the alkyl group has 1 to 10 carbons, or a cycloalkyl group of 5 to 10 carbons, or R 8 is phenyl or lower alkylphenyl
  • X is O, S, or (CR 1 R 1 ) n where n is 0, 1 or 2; Y is a bivalent radical having Formulae XXII or XXIII where o is an integer between 1 through 4
  • Y is a bivalent aryl or 5 or 6 membered heteroaryl radical having 1 to 3 heteroatoms selected from N, S and O, the aryl or heteroaryl groups being unsubstituted, or substituted with 1 to 3 C 1-6 alkyl or with 1 to 3 C 1-6 fluoroalkyl groups with the proviso that when the compound is in accordance with Formula II then Y is not a 5 or 6 membered ring;
  • X 1 is S or NH;
  • R 1 is independently H, lower alkyl of 1 to 6 carbons, or lower fluoroalkyl of 1 to 6 carbons;
  • R 2 is independently H, lower alkyl of 1 to 6 carbons, OR 1 , adamantly, or lower fluoroalkyl of 1 to 6 carbons, or the two R 2 groups jointly represent an oxo ( ⁇ O) group with the proviso that when the compound is in accordance with Formula II then at least one of the R 2 substituents is branched-chain al
  • R is H, lower alkyl of 1 to 6 carbons, or a pharmaceutically acceptable salt of the compound.
  • R is H, lower alkyl of 1 to 6 carbons, and R 1 is iso-propyl or tertiary-butyl, or a pharmaceutically acceptable salt of the compound.
  • R is H, lower alkyl of 1 to 6 carbons, and R 1 is iso-propyl, n-butyl or tertiary-butyl, or a pharmaceutically acceptable salt of the compound.
  • X is O or S
  • Y is a bivalent cycloalkyl or cycloalkenyl radical optionally substituted with one to four R 4 groups, the cycloalkenyl radical having 5 to 6 carbons and one double bond, or Y is a bivalent aryl or 5 or 6 membered heteroaryl radical having 1 to 3 heteroatoms selected from N, S and O, the aryl or heteroaryl groups optionally substituted with 1 to 4 R 4 groups with the proviso that the cycloalkyl or the cycloalkenyl radical is not substituted on the same carbon with the condensed cyclic moiety and with the diene containing moiety;
  • R 1 is independently H, alkyl of 1 to 6 carbons, or fluoroalkyl of 1 to 6 carbons;
  • R 2 is independently H, alkyl of 1 to 8 carbons, or fluoroalkyl of 1 to 8 carbons;
  • R 12 is independently H, alkyl of 1 to 8 carbons, or
  • R 4 is H, halogen, alkyl of 1 to 10 carbons, fluoro substituted alkyl of 1 to 6 carbons, alkoxy of 1 to 10 carbons, or alkylthio of 1 to 10 carbons; m is an integer having the values of 0 to 3; r is an integer having the values of 1 to 10; s is an integer having the values 1 to 4; t is an integer having the values 1 to 5;
  • R 7 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons
  • R 8 is an alkyl group of 1 to 10 carbons, a cycloalkyl group of 5 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or R 8 is phenyl or lower alkylphenyl, R 9 and R 10 independently are
  • R 1 is H or methyl
  • R 8 is H, alkyl of 1 to 6 carbons, or a pharmaceutically acceptable cation
  • R 3 is hydrogen, alkyl of 1 to 10 carbons, halogen, alkoxy of 1 to 10 carbons, or R 3 is selected from the groups shown below
  • R 4 is H, halogen, alkyl of 1 to 10 carbons, carbons, alkoxy of 1 to 10; r is an integer having the values of 1 to 10; s is an integer having the values 1 to 4;
  • t represents a 5 or 6 membered heteroaryl ring having 1 to 3 heteroatoms selected from the group consisting of N, S and O, and t is an integer having the values 1 to 5.
  • a RXR agonist can induce Treg cell differentiation
  • the ability of an RXR agonist to promote Treg cell differentiation under Treg cell differentiation conditions was assessed by monitoring Foxp3 and ⁇ 4 ⁇ 7 expression.
  • Naive CD4′ CD25 ⁇ FoxP3 ⁇ cells were purified from a Foxp3-GFP mouse using flow cytometry by sorting and isolating based upon a GFP ⁇ phenotype. These cells were then cultured under Treg cell differentiation conditions by treating the cells with ⁇ CD3 and ⁇ CD28 polyclonal antibodies in the presence of IL-2 and TGF- ⁇ .
  • the cultured cells were incubated with RXR agonist 194204 (Formula XXIX) at 0.1 nM, 1.0 nM and 10 nM and the expression of Foxp3 and ⁇ 4 ⁇ 7 was analyzed.
  • the results indicate that RXR agonist exerted significant impact on the expression of Foxp3, inducing nearly 100% Foxp3 T cells at concentrations of 1 nM or higher.
  • FIG. 1A These results also indicate that RXR agonist 194204 also induced expression of ⁇ 4 ⁇ 7 (a gut homing receptor).
  • FIG. 1B These results indicate that RXR agonists could be useful in reducing a symptom of an autoimmune disorder or a transplant rejection.
  • a RXR agonist can regulate T cell differentiation.
  • the ability of an RXR agonist to promote Treg cell differentiation and inhibit Th17 cell differentiation under Th17 cell differentiation conditions was assessed by monitoring Foxp3 and IL-17A expression.
  • Naive CD4′ CD25 ⁇ FoxP3 ⁇ cells were purified from a Foxp3-GFP mouse using flow cytometry by sorting and isolating based upon a GFP ⁇ phenotype. These cells were then cultured under Th17 cell differentiation conditions in media with 0 nM, 1 nM, 10 nM, and 100 nM of RXR agonist 194204 (Formula XXIX) and the expression of Foxp3 and IL-17A was analyzed.
  • RXR Agonists Regulate T Cell Differentiation Independent of RAR Signaling
  • T cells were incubated with a RXR agonist in the presence of a pan-RAR antagonist and the expression of Foxp3 was assessed.
  • Naive CD4′ CD25 ⁇ FoxP3 ⁇ cells were purified from a Foxp3-GFP mouse using flow cytometry by sorting and isolating based upon a GFP ⁇ phenotype. These cells were then cultured under Treg cell differentiation conditions by treating the cells with ⁇ CD3 and ⁇ CD28 polyclonal antibodies in the presence of IL-2 and TGF- ⁇ .
  • the cultured cells were incubated with RXR agonist 194204 (Formula XXIX) at 1.0 nM together with 0 nM, 1 nM, or 10 nM of a pan-RAR antagonist 194310.
  • the cultured cells were then assayed for the expression of Foxp3.
  • the results indicate that the inclusion of a pan-RAR antagonist only partially blocked the induction of Foxp3 expression observed with an RXR agonist alone.
  • FIG. 3 shows that this partial inhibition of Fox3p expression may actually be due to the blocking of the effects of endogenous RA in the culture medium.
  • these results indicate that the observed conversion of T cells into Treg cells appears to occur through the use of RXR receptor homodimers and/or some other RXR containing complex, and not through a RAR-mediated mechanism.
  • RXR ⁇ receptor homodimers To determine whether a RXR agonist can mediate its effects via an RXR ⁇ receptor homodimers, RXR ⁇ receptor homodimers, RXR ⁇ receptor homodimers, or any combination thereof, or the corresponding RAR/RXR heterodimers, receptor-mediated transactivation assays were performed.
  • transactivation assays assessing RXR homodimer signaling CV-1 cells were transfected with 1) an expression construct including a full length RXR ⁇ , RXR ⁇ , or RXR ⁇ ; and 2) a rCRBPII/RXRE-tk-Luc reporter construct that included RXR homodimer-specific RXRE/DR1 responsive element linked to a luciferase gene.
  • CV-1 cells were transfected with 1) an expression construct comprising a fusion protein including an estrogen receptor (ER) DNA binding domain linked to the ligand binding domain of RAR ⁇ , RAR ⁇ , or RAR ⁇ and 2) a ERE-tk-Luc reporter construct that included an estrogen receptor responsive element linked to a luciferase gene.
  • ER estrogen receptor
  • the ER-RAR fusion proteins provided an accurate readout of only the transfected ER-RAR.
  • CV-1 cells were treated with RXR agonist 194204 (Formula XXIX) at increasing concentrations for 20 hours before measuring luciferase activity.
  • Luciferase activity is expressed as percent of maximal activity obtained using 1 ⁇ M RXR agonist 194204 for RXRs and 1 ⁇ M all-trans-retinoic acid (ATRA) for RARs (Table 1). Data are mean values ⁇ SE from five independent experiments.
  • RXR agonist 194204 activated RXR receptors with very high potency (EC 50 ⁇ 0.5 nM) for all three RXR subtypes (Table 1).
  • EC 50 of the RXR agonist for RARs was >1,000 nM with minimal activity detected at ⁇ 1 ⁇ M. This difference represents >2.000-fold selectivity for RXRs over RARs in functional transactivation assays.
  • RXR agonist 194204 was more than 1.000-fold more potent in activating RXR receptors rather than RAR receptors.
  • FIG. 4 These results indicate that Treg differentiation was mediated through a RXR signaling pathway and not via a RAR signaling pathway.
  • RXR agonist 194204 was shown not to transactivate so called “permissive RXR heterodimers”, such as, e.g., PPAR/RXR, FXR/RXR and LXR/RXR.
  • RXR agonist 194204 is distinct from other RXR agonists.
  • RXR ⁇ , RXR ⁇ , RXR ⁇ , RAR ⁇ , RAR ⁇ , or RAR ⁇ were expressed in SF21 cells using a baclovirus expression system and the resulting proteins were purified.
  • purified RXR ⁇ , RXR ⁇ , and RXR ⁇ were separately incubated with 10 nM [ 3 H]-9CRA, and the binding affinity of the RXR agonist 194204 (Formula XXIX) was determined by competitive displacement of [ 3 H]-9CRA from the receptor.
  • RXR agonist 194204 displayed high affinity for RXR ⁇ , RXR ⁇ , and RXR ⁇ with Ki values being 1.7, 16, and 43 nM, respectively.
  • the RXR agonist 194204 bound with very low affinity to each of the RARs (Ki values being >1,000 nM).
  • C57BL/6 (B6) mice were immunized (day 0) to induce EAE by subcutaneous (s.c.) injection at the base of their spine with 200 uL of adjuvant containing 125 ug myelin oligodendrocyte glycoprotein peptide (35-55) (MOG peptide; Peptides International, Louisville, Ky.) and 400 ug non-viable M. tuberculosis H37 desiccate emulsified in a mixture of incomplete Freund's adjuvant and phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • mice were scored using the following scale: 0—Mice have no disease, 1—Mice have distal limp tail or rear leg weakness (paresis), 1.5—Mice have distal limp tail and rear leg weakness, 2—Mice have complete limp tail and rear leg weakness, 2.5—Mice have complete limp tail and weakness in both rear legs, 3—Mice have complete limp tail and paralysis in both rear legs, 3.5—Mice have complete limp tail, paralysis in both rear legs, and forelimb weakness. Mice receiving a score of 3.5 were immediately euthanized.
  • FIG. 5 depicts scores of disease severity over time. The results indicate that administration of a RXR agonist significantly reduces the symptoms of EAE in mice. Efficacy of the RXR agonist was observed after the first administration (day 7) and maintained throughout the course of the study (day 20).
  • CNS infiltrating cells C57BL/6 (B6) mice were treated as described in Example 6. On day 20 after immunization, mice were sacrificed and perfused with phosphate buffered saline (PBS). Brain and spinal cord tissue was isolated, digested with DNase and Liberase DL (Roche Diagnostics, Indianapolis, Ind.) for 30 minutes, and homogenized through 70 micron nylon mesh filters. Resulting cells were placed over a Percoll gradient to remove myelin. The remaining cells (microglia and CNS infiltrating cells) were counted, stained for molecules of interest, and run on a flow cytometer. Based on the frequencies obtained by FACS of these cell populations, total cell numbers of CNS infiltrating leukocytes expressing CD45, including CD4′ T cells and CD11c + CD11b + myeloid dendritic cells (DC), were calculated.
  • DC central nervous system
  • FIG. 6 compares the number of CD4 + cells or CD11c + CD11b + cells (myeloid DC) in mice treated with the RXR agonist 194204 verses the vehicle control. There was a significant reduction in the infiltration of both CD4 + cells and CD11c + CD11b + cells in animals treated with a RXR agonist as compared to the control. As disease is propagated in the CNS through the CD4 + cells infiltrating the CNS and becoming re-activated by CD11c + CD11b + cells, this suggests that part of the mechanism of action in this model is to limit the presence of the cells in the CNS.
  • mice were immunized to induce EAE by s.c. injection at the base of their spine with 200 uL of adjuvant containing 200 ug proteolipid proteins (139-151) (PLP peptide; Peptides International, Louisville, Ky.) and 400 ug of non-viable M. tuberculosis H37 desiccate emulsified in a mixture of incomplete Freund's adjuvant and PBS.
  • FIG. 7 depicts scores of disease severity over time. Efficacy of the RXR agonist was observed after the second administration (day 8) and maintained throughout the course of the study (day 14).
US13/714,051 2011-12-13 2012-12-13 Autoimmune disorder treatment using rxr agonists Abandoned US20130190395A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
US13/714,051 US20130190395A1 (en) 2011-12-13 2012-12-13 Autoimmune disorder treatment using rxr agonists
US14/507,730 US10653650B2 (en) 2011-12-13 2014-10-06 Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective RXR agonists
US14/626,339 US10034845B2 (en) 2011-12-13 2015-02-19 Autoimmune disorder treatment using RXR agonists
US14/732,328 US20150342917A1 (en) 2011-12-13 2015-06-05 Autoimmune Disorder Treatment Using RXR Agonists
US15/341,969 US10285960B2 (en) 2011-12-13 2016-11-02 Autoimmune disorder treatment using RXR agonists
US15/852,580 US10201512B2 (en) 2011-12-13 2017-12-22 Autoimmune disorder treatment using RXR agonists
US15/988,965 US10695307B2 (en) 2011-12-13 2018-05-24 Autoimmune disorder treatment using RXR agonists
US16/228,217 US10945976B2 (en) 2011-12-13 2018-12-20 Autoimmune disorder treatment using RXR agonists
US16/736,705 US20200163915A1 (en) 2011-12-13 2020-01-07 Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective rxr agonists
US16/742,616 US11547684B2 (en) 2011-12-13 2020-01-14 Autoimmune disorder treatment using RXR agonists
US16/742,600 US20200155488A1 (en) 2011-12-13 2020-01-14 Autoimmune disorder treatment using rxr agonists
US17/126,787 US11246845B2 (en) 2011-12-13 2020-12-18 Autoimmune disorder treatment using RXR agonists
US17/126,714 US11166927B2 (en) 2011-12-13 2020-12-18 Autoimmune disorder treatment using RXR agonists
US17/560,035 US11576881B2 (en) 2011-12-13 2021-12-22 Autoimmune disorder treatment using RXR agonists
US17/586,657 US11793781B2 (en) 2011-12-13 2022-01-27 Autoimmune disorder treatment using RXR agonists
US17/859,743 US20220370386A1 (en) 2011-12-13 2022-07-07 Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective rxr agonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161570182P 2011-12-13 2011-12-13
US13/714,051 US20130190395A1 (en) 2011-12-13 2012-12-13 Autoimmune disorder treatment using rxr agonists

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US14/507,730 Continuation-In-Part US10653650B2 (en) 2011-12-13 2014-10-06 Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective RXR agonists
US14/626,339 Continuation US10034845B2 (en) 2011-12-13 2015-02-19 Autoimmune disorder treatment using RXR agonists
US14/732,328 Division US20150342917A1 (en) 2011-12-13 2015-06-05 Autoimmune Disorder Treatment Using RXR Agonists
US15/341,969 Continuation US10285960B2 (en) 2011-12-13 2016-11-02 Autoimmune disorder treatment using RXR agonists

Publications (1)

Publication Number Publication Date
US20130190395A1 true US20130190395A1 (en) 2013-07-25

Family

ID=48613185

Family Applications (13)

Application Number Title Priority Date Filing Date
US13/714,051 Abandoned US20130190395A1 (en) 2011-12-13 2012-12-13 Autoimmune disorder treatment using rxr agonists
US14/626,339 Active US10034845B2 (en) 2011-12-13 2015-02-19 Autoimmune disorder treatment using RXR agonists
US14/732,328 Pending US20150342917A1 (en) 2011-12-13 2015-06-05 Autoimmune Disorder Treatment Using RXR Agonists
US15/341,969 Active US10285960B2 (en) 2011-12-13 2016-11-02 Autoimmune disorder treatment using RXR agonists
US15/852,580 Active US10201512B2 (en) 2011-12-13 2017-12-22 Autoimmune disorder treatment using RXR agonists
US15/988,965 Active US10695307B2 (en) 2011-12-13 2018-05-24 Autoimmune disorder treatment using RXR agonists
US16/228,217 Active US10945976B2 (en) 2011-12-13 2018-12-20 Autoimmune disorder treatment using RXR agonists
US16/742,616 Active 2033-07-21 US11547684B2 (en) 2011-12-13 2020-01-14 Autoimmune disorder treatment using RXR agonists
US16/742,600 Abandoned US20200155488A1 (en) 2011-12-13 2020-01-14 Autoimmune disorder treatment using rxr agonists
US17/126,787 Active US11246845B2 (en) 2011-12-13 2020-12-18 Autoimmune disorder treatment using RXR agonists
US17/126,714 Active US11166927B2 (en) 2011-12-13 2020-12-18 Autoimmune disorder treatment using RXR agonists
US17/560,035 Active US11576881B2 (en) 2011-12-13 2021-12-22 Autoimmune disorder treatment using RXR agonists
US17/586,657 Active US11793781B2 (en) 2011-12-13 2022-01-27 Autoimmune disorder treatment using RXR agonists

Family Applications After (12)

Application Number Title Priority Date Filing Date
US14/626,339 Active US10034845B2 (en) 2011-12-13 2015-02-19 Autoimmune disorder treatment using RXR agonists
US14/732,328 Pending US20150342917A1 (en) 2011-12-13 2015-06-05 Autoimmune Disorder Treatment Using RXR Agonists
US15/341,969 Active US10285960B2 (en) 2011-12-13 2016-11-02 Autoimmune disorder treatment using RXR agonists
US15/852,580 Active US10201512B2 (en) 2011-12-13 2017-12-22 Autoimmune disorder treatment using RXR agonists
US15/988,965 Active US10695307B2 (en) 2011-12-13 2018-05-24 Autoimmune disorder treatment using RXR agonists
US16/228,217 Active US10945976B2 (en) 2011-12-13 2018-12-20 Autoimmune disorder treatment using RXR agonists
US16/742,616 Active 2033-07-21 US11547684B2 (en) 2011-12-13 2020-01-14 Autoimmune disorder treatment using RXR agonists
US16/742,600 Abandoned US20200155488A1 (en) 2011-12-13 2020-01-14 Autoimmune disorder treatment using rxr agonists
US17/126,787 Active US11246845B2 (en) 2011-12-13 2020-12-18 Autoimmune disorder treatment using RXR agonists
US17/126,714 Active US11166927B2 (en) 2011-12-13 2020-12-18 Autoimmune disorder treatment using RXR agonists
US17/560,035 Active US11576881B2 (en) 2011-12-13 2021-12-22 Autoimmune disorder treatment using RXR agonists
US17/586,657 Active US11793781B2 (en) 2011-12-13 2022-01-27 Autoimmune disorder treatment using RXR agonists

Country Status (7)

Country Link
US (13) US20130190395A1 (zh)
EP (1) EP2790698A1 (zh)
CN (2) CN110151745A (zh)
AU (2) AU2012352149B2 (zh)
CA (1) CA2858882C (zh)
MX (1) MX352727B (zh)
WO (1) WO2013090616A1 (zh)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017155577A1 (en) 2016-03-10 2017-09-14 Io Therapeutics, Inc. Treatment of autoimmune diseases with combinations of rxr agonists and thyroid hormones
US9877941B2 (en) 2015-10-31 2018-01-30 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10034845B2 (en) 2011-12-13 2018-07-31 lo Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
JP2019511492A (ja) * 2016-03-10 2019-04-25 アイオー セラピューティクス インコーポレイテッド Rxrアゴニストおよび甲状腺ホルモンの組み合わせでの筋肉障害の治療
US10285985B2 (en) 2015-03-31 2019-05-14 National University Corporation Okayama University Pharmaceutical composition for treatment or prevention of neurodegenerative diseases
US10590059B2 (en) 2017-11-17 2020-03-17 Io Therapeutics, Inc. Compounds and synthetic methods for the preparation of retinoid X receptor-specific retinoids
US10596133B2 (en) 2005-09-30 2020-03-24 Io Therapeutics, Inc. Treatment of cancer with specific RXR agonists
US10653650B2 (en) 2011-12-13 2020-05-19 Io Therapeutics, Inc. Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective RXR agonists
US10966950B2 (en) 2019-06-11 2021-04-06 Io Therapeutics, Inc. Use of an RXR agonist in treating HER2+ cancers
US11517549B2 (en) 2017-09-20 2022-12-06 Io Therapeutics, Inc. Treatment of disease with esters of selective RXR agonists
US11896558B2 (en) 2021-12-07 2024-02-13 Io Therapeutics, Inc. Use of an RXR agonist and taxanes in treating Her2+ cancers

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3210926B1 (en) * 2016-02-23 2020-01-01 Otis Elevator Company Elevator service panel
MA54296A (fr) 2018-11-26 2021-10-06 Denali Therapeutics Inc Procédés de traitement du métabolisme lipidique dérégulé
EP3977797A4 (en) * 2019-05-31 2022-12-28 Qualcomm Incorporated PARTIAL RECIPROCITY CSI DETECTION

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5917082A (en) * 1995-06-06 1999-06-29 Allergan Sales, Inc. 2,4-pentadienoic acid derivatives having retinoid-like biological activity
US20070185055A1 (en) * 2006-02-06 2007-08-09 Guang Liang Jiang Method for treating cachexia with retinoid ligands

Family Cites Families (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4756911A (en) 1986-04-16 1988-07-12 E. R. Squibb & Sons, Inc. Controlled release formulation
US5378475A (en) 1991-02-21 1995-01-03 University Of Kentucky Research Foundation Sustained release drug delivery devices
JPH08506323A (ja) 1992-11-25 1996-07-09 ラ ホヤ キャンサー リサーチ ファウンデーション Rxrホモダイマー形成ならびに架橋二環式芳香族化合物および調節遺伝子発現におけるそれらの使用
US5466861A (en) 1992-11-25 1995-11-14 Sri International Bridged bicyclic aromatic compounds and their use in modulating gene expression of retinoid receptors
WO1994014777A1 (en) 1992-12-28 1994-07-07 Eisai Co., Ltd. Heterocyclic carbonic acid derivatives which bind to retinoid receptors (rar)
US5455265A (en) 1993-02-11 1995-10-03 Allergan, Inc. Method of treatment with compounds having selective agonist-like activity on RXR retinoid receptors
US5869079A (en) 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
US5675033A (en) 1995-06-06 1997-10-07 Allergan 2,4-pentadienoic acid derivatives having retinoid-like biological activity
JP3964478B2 (ja) 1995-06-30 2007-08-22 エーザイ・アール・アンド・ディー・マネジメント株式会社 ヘテロ環含有カルボン酸誘導体及びそれを含有する医薬
US5952345A (en) 1995-09-01 1999-09-14 Allergan Sales, Inc. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US6218128B1 (en) 1997-09-12 2001-04-17 Allergan Sales, Inc. Methods of identifying compounds having nuclear receptor negative hormone and/or antagonist activities
US6008204A (en) 1995-09-01 1999-12-28 Allergan Sales, Inc. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US6942980B1 (en) 1995-09-01 2005-09-13 Allergan, Inc. Methods of identifying compounds having nuclear receptor negative hormone and/or antagonist activities
US5958954A (en) 1995-09-01 1999-09-28 Allergan Sales, Inc. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US5776699A (en) 1995-09-01 1998-07-07 Allergan, Inc. Method of identifying negative hormone and/or antagonist activities
NZ319158A (en) 1995-10-02 1999-10-28 Hoffmann La Roche Pyrimidine derivatives as 5ht2c-receptor antagonists
US5675024A (en) 1995-11-22 1997-10-07 Allergan Aryl or heteroaryl amides of tetrahydronaphthalene, chroman, thiochroman and 1,2,3,4,-tetrahydroquinoline carboxylic acids, having an electron withdrawing substituent in the aromatic or heteroaromatic moiety, having retinoid-like biological activity
US5965606A (en) 1995-12-29 1999-10-12 Allergan Sales, Inc. Methods of treatment with compounds having RAR.sub.α receptor specific or selective activity
US5877207A (en) 1996-03-11 1999-03-02 Allergan Sales, Inc. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US20030219832A1 (en) 1996-03-11 2003-11-27 Klein Elliott S. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US6441025B2 (en) 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US5763635A (en) 1996-06-21 1998-06-09 Allergan Tetrahydronaphthalene derivatives substituted in the 8 position with alkyhidene groups having retinoid and/or retinoid antagonist-like biological activity
US5773594A (en) 1996-06-21 1998-06-30 Allergan Alkyl or aryl substituted dihydronaphthalene derivatives having retinoid and/or retinoid antagonist-like biological activity
US6555690B2 (en) 1996-06-21 2003-04-29 Allergan, Inc. Alkyl or aryl substituted dihydronaphthalene derivatives having retinoid and/or retinoid antagonist-like biological activity
US5739338A (en) 1996-11-05 1998-04-14 Allergan N-aryl substituted tetrahydroquinolines having retinoid agonist, retinoid antagonist or retinoid inverse agonist type biological activity
DE69726182T2 (de) 1996-12-11 2004-08-12 Dana-Farber Cancer Institute, Inc., Boston Methoden und pharmazeutische zusammenstellungen zur wachstumsverhinderung von tumorzellen enthaltend einen ppar-gamma agonisten und einen map kinase inhibitor
US5728846A (en) 1996-12-12 1998-03-17 Allergan Benzo 1,2-g!-chrom-3-ene and benzo 1,2-g!-thiochrom-3-ene derivatives
US6037488A (en) 1997-04-19 2000-03-14 Allergan Sales, Inc. Trisubstituted phenyl derivatives having retinoid agonist, antagonist or inverse agonist type biological activity
US5919970A (en) 1997-04-24 1999-07-06 Allergan Sales, Inc. Substituted diaryl or diheteroaryl methanes, ethers and amines having retinoid agonist, antagonist or inverse agonist type biological activity
US6670398B2 (en) 1997-05-14 2003-12-30 Atherogenics, Inc. Compounds and methods for treating transplant rejection
US6063768A (en) 1997-09-04 2000-05-16 First; Eric R. Application of botulinum toxin to the management of neurogenic inflammatory disorders
EP1077919A1 (en) 1998-05-11 2001-02-28 Novo Nordisk A/S New compounds, their preparation and use
EP1093362A1 (en) 1998-06-12 2001-04-25 Ligand Pharmaceuticals Incorporated Treatment of anti-estrogen resistant breast cancer using rxr modulators
US6403638B1 (en) 1998-10-01 2002-06-11 Allergan Sales, Inc. 2,4-pentadienoic acid derivatives having selective activity for retinoid X (RXR) receptors
US6147224A (en) 1998-10-01 2000-11-14 Allergan Sales, Inc. 2,4-pentadienoic acid derivatives having selective activity for retinoid X (RXR) receptors
US6048873A (en) 1998-10-01 2000-04-11 Allergan Sales, Inc. Tetrahdroquinolin-2-one 6 or 7-yl, tetrahdroquinilin-2-thione 6 or 7-yl pentadienoic acid and related derivatives having retinoid-like biological activity
US6521641B1 (en) 1998-10-08 2003-02-18 Allergan, Inc. Male anti-fertility agents
US6776984B1 (en) 1999-08-20 2004-08-17 George R. Schwartz Induced regeneration and repair of damaged neurons and nerve axon myelin
US6043381A (en) 1999-05-07 2000-03-28 Allergan Sales, Inc. Process for preparing substituted benzo[1,2-g]-chrom-3-ene, benzo[1,2-g]-thiochrom-3-ene and benzo[1,2-g]-1,2-dihydroquinoline derivatives
WO2001007028A2 (en) 1999-07-23 2001-02-01 Allergan Sales, Inc. The use of retinoid receptor antagonists in the treatment of prostate carcinoma
US6187750B1 (en) 1999-08-25 2001-02-13 Everyoung Technologies, Inc. Method of hormone treatment for patients with symptoms consistent with multiple sclerosis
EP1216221A2 (en) 1999-09-14 2002-06-26 Ligand Pharmaceuticals Incorporated Rxr modulators with improved pharmacologic profile
US6313168B1 (en) 1999-12-15 2001-11-06 Allergan Sales, Inc. Use of retinoid receptor antagonists in the treatment of cartilage and bone pathologies
TWI281911B (en) 2000-04-04 2007-06-01 Allergan Inc Treatment of tumors with RARalpha selective retinoid compounds in combination with other anti-tumor agents
US20030077664A1 (en) 2001-04-18 2003-04-24 Yi Zhao Methods of screening for compounds that modulate hormone receptor activity
US20020193403A1 (en) 2001-05-03 2002-12-19 Allergan Sales, Inc. Methods of treating hyperlipidemia
SK14632003A3 (sk) 2001-05-08 2004-03-02 Merck Patent Gmbh Kombinovaná terapia pri použití anti-EGFR protilátok a antihormonálnych činidiel
US7173134B2 (en) 2001-09-25 2007-02-06 Smithkline Beecham Corporation Selective RXR ligands
US6645526B2 (en) 2001-11-13 2003-11-11 Mylan Pharmaceuticals, Inc. Storage stable thyroxine active drug formulations and methods for their production
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
US20050220774A1 (en) 2002-03-18 2005-10-06 Tony Peled Methods of inducing differentiation in ex vivo expanded stem cells
US6720423B2 (en) 2002-04-30 2004-04-13 Allergan, Inc. Dihydrobenzofuran and dihydrobenzothiophene 2,4-pentadienoic acid derivatives having selective activity for retinoid X (RXR) receptors
PL372556A1 (en) 2002-06-04 2005-07-25 Galderma Research & Development, S.N.C. Novel ligands that are inhibitors of the rar receptors, process for preparing them and use thereof in human medicine and in cosmetics
US20040005304A1 (en) 2002-07-08 2004-01-08 Mak Wood, Inc. Novel compositions and methods for treating neurological disorders and associated gastrointestinal conditions
JP4275365B2 (ja) 2002-08-06 2009-06-10 三菱電機株式会社 複合形ガス絶縁開閉装置
US7105566B2 (en) 2002-10-22 2006-09-12 Allergan, Inc. Methods of treatment during vascular procedures
ES2283866T3 (es) 2002-11-18 2007-11-01 GALDERMA RESEARCH & DEVELOPMENT Nuevos ligandos que son antagonistas de los receptores rar, procedimiento de preparacion de estos ligandos y utilizacion de estos ligandos en el campo de la medicina humana y de la cosmetica.
US7019034B2 (en) 2003-01-28 2006-03-28 Allergan, Inc. Compositions and methods for reducing serum glucose and triglyceride levels in diabetic mammals
US6936636B2 (en) 2003-06-26 2005-08-30 Allergan, Inc. 5-[phenyl-tetrahydronaphthalene-2-yl dihydronaphthalen-2-yl and heteroaryl-cyclopropyl]-pentadienoic acid derivatives having serum glucose reducing activity
CA2535260A1 (en) 2003-08-07 2005-02-17 Allergan, Inc. Method for treating cachexia with retinoid ligands
CA2539288C (en) 2003-09-15 2015-05-12 Shaker A. Mousa Thyroid hormone analogs and methods of use
WO2005046726A2 (en) * 2003-11-12 2005-05-26 Allergan, Inc. Combinations for inhibiting cell growth which contains an inhibitor of human ck1 alpha activity and a rxr agonist
CA2553381C (en) 2004-01-20 2011-03-22 Allergan, Inc. Compositions for localized therapy of the eye, comprising preferably triamcinolone acetonide and hyaluronic acid
US7799336B2 (en) 2004-04-30 2010-09-21 Allergan, Inc. Hypotensive lipid-containing biodegradable intraocular implants and related methods
WO2006030442A2 (en) 2004-09-16 2006-03-23 Gamida-Cell Ltd. Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
US8105611B2 (en) 2005-06-17 2012-01-31 Allergan, Inc. Treatment of autoimmune disorder with a neurotoxin
CA2617623A1 (en) 2005-08-18 2007-02-22 Victoria M. Richon Combination methods of saha and targretin for treating cancer
SI1937244T1 (sl) 2005-09-30 2018-12-31 Io Therapeutics, Llc Zdravljenje raka s specifičnimi RXR agonisti
US7723326B2 (en) * 2005-09-30 2010-05-25 Janssen Pharmaceutica N.V. Heterocyclic amide derivatives as RXR agonists for the treatment of dyslipidemia, hypercholesterolemia and diabetes
CA2624345A1 (en) * 2005-09-30 2007-04-12 Janssen Pharmaceutica N.V. 1,2,3,5-tetrahydro-cyclopental[c]quinolin-4-one derivatives as rxr agonists for the treatment of dyslipidemia, hypercholesterolemia and diabetes
WO2008109445A1 (en) 2007-03-02 2008-09-12 Preventive Nutrient Company, Inc. Compositions and methods for treating alzheimer's disease and dementia
JP2010531138A (ja) * 2007-06-13 2010-09-24 ラ ホヤ インスティテュート フォア アラージー アンド イムノロジー 制御性t細胞ならびに同製造および使用方法
WO2009102789A2 (en) 2008-02-15 2009-08-20 Wyeth Use of rxr agonists for the treatment of osteroarthritis
ES2332169B1 (es) 2008-07-24 2010-10-25 Universidad Del Pais Vasco Empleo de microparticulas que comprenden celulas modificadas geneticamente en el tratamiento de enfermedades neurodegenerativas.
BRPI0920546B1 (pt) * 2008-10-08 2021-08-10 Cambridge Enterprise Limited Métodos para identificar e selecionar um paciente com esclerose múltipla
JP5542059B2 (ja) 2008-10-09 2014-07-09 国立大学法人 岡山大学 Rxr作動性物質を有効成分とする抗アレルギー剤
WO2010123945A2 (en) 2009-04-20 2010-10-28 Allergan, Inc. Silk fibroin hydrogels and uses thereof
US20100292281A1 (en) 2009-05-15 2010-11-18 The University Of Kentucky Research Foundation Treatment of mci and alzheimer's disease
US20100298434A1 (en) 2009-05-21 2010-11-25 Claude Rouillard Neuroprotection and prevention of dopaminergic cell death by targeting nur77 translocation
JP2010280585A (ja) * 2009-06-02 2010-12-16 Okayama Univ Rxr作動性物質を有効成分とする鎮痛剤
EP2451455A4 (en) * 2009-07-10 2013-01-16 Univ Case Western Reserve RXR AGONIST COMPOUNDS AND METHODS
AU2011218019B2 (en) 2010-02-19 2016-03-24 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona, Acting For And On Behalf Of Arizona State University Novel bexarotene analogs
WO2012125749A2 (en) 2011-03-14 2012-09-20 Io Therapeutics, Inc. INFLAMMATION AND AUTOIMMUNE DISORDER TREATMENT USING RARα SELECTIVE AGONISTS
EP2556827A1 (en) 2011-08-11 2013-02-13 Acadia Pharmaceuticals Inc. Treatment of neurodegenerative diseases
EP2766018A4 (en) 2011-10-13 2015-02-25 Univ Case Western Reserve RXR AGONIST COMPOUNDS AND ASSOCIATED METHODS
WO2013059632A1 (en) 2011-10-19 2013-04-25 John Rankin Method for indirect food temperature measurement
US10653650B2 (en) 2011-12-13 2020-05-19 Io Therapeutics, Inc. Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective RXR agonists
MX352727B (es) 2011-12-13 2017-12-06 Dartmouth College Tratamiento de trastorno autoinmune al usar agonistas de rxr.
US9446105B2 (en) 2013-03-15 2016-09-20 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor specific for folate receptor β
US20160263189A1 (en) 2013-10-23 2016-09-15 Acadia Pharmaceuticals Inc. Treatment of a neurodegenerative disease or disorder
WO2015066197A2 (en) 2013-10-29 2015-05-07 Vestion, Inc. Cardiac neural crest cells and methods of use thereof
JP2018512396A (ja) 2015-03-09 2018-05-17 キングス・カレッジ・ロンドン Th1応答を増強するためのrarアルファアゴニストを用いた併用療法
WO2017075612A1 (en) 2015-10-31 2017-05-04 Io Therapeutics, Inc. Treatment of cancer with combinations of rxr agonists and thyroid hormones
PL3368080T3 (pl) 2015-10-31 2023-09-11 Io Therapeutics, Inc. Leczenie zaburzeń układu nerwowego z zastosowaniem kombinacji agonistów rxr i hormonów tarczycy
EP3368160B1 (en) 2015-10-31 2023-05-17 IO Therapeutics, Inc. Treatment of nervous system disorders using thyroid hormone neutral doses of rxr agonists
KR20230164204A (ko) 2016-03-10 2023-12-01 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
EP3426303B1 (en) 2016-03-10 2022-06-15 IO Therapeutics, Inc. Treatment of muscular disorders with combinations of rxr agonists and thyroid hormones
AU2018326617B2 (en) 2017-08-31 2022-12-01 Io Therapeutics, Inc. Rar selective agonists in combination with immune modulators for cancer immunotherapy
CA3076373A1 (en) 2017-09-20 2019-03-28 Io Therapeutics, Inc. Treatment of disease with esters of selective rxr agonists
JP2021503464A (ja) 2017-11-17 2021-02-12 アイオー セラピューティクス インコーポレイテッド レチノイドx受容体特異的なレチノイドの調製のための化合物および合成方法
US10966950B2 (en) 2019-06-11 2021-04-06 Io Therapeutics, Inc. Use of an RXR agonist in treating HER2+ cancers

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5917082A (en) * 1995-06-06 1999-06-29 Allergan Sales, Inc. 2,4-pentadienoic acid derivatives having retinoid-like biological activity
US20070185055A1 (en) * 2006-02-06 2007-08-09 Guang Liang Jiang Method for treating cachexia with retinoid ligands

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Beyer et al. Weight change and body composition in patients with Parkinson's disease.J AM. DietAssoc. (1995), vol. 95, pp.979-983. *
Levesque et al. Nur77 and retinoid X receptors: crucial factors in dopamine-related neuroadaptation. Trends in Neuroscience (2007) vol .30, pp. 22-30. *

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10973788B2 (en) 2005-09-30 2021-04-13 Io Therapeutics, Inc. Treatment of cancer with specific RXR agonists
US10596133B2 (en) 2005-09-30 2020-03-24 Io Therapeutics, Inc. Treatment of cancer with specific RXR agonists
US11576881B2 (en) 2011-12-13 2023-02-14 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10034845B2 (en) 2011-12-13 2018-07-31 lo Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US11793781B2 (en) 2011-12-13 2023-10-24 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10201512B2 (en) 2011-12-13 2019-02-12 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10945976B2 (en) 2011-12-13 2021-03-16 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US11547684B2 (en) 2011-12-13 2023-01-10 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10695307B2 (en) 2011-12-13 2020-06-30 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10285960B2 (en) 2011-12-13 2019-05-14 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US11246845B2 (en) 2011-12-13 2022-02-15 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10653650B2 (en) 2011-12-13 2020-05-19 Io Therapeutics, Inc. Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective RXR agonists
US11166927B2 (en) 2011-12-13 2021-11-09 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
US10285985B2 (en) 2015-03-31 2019-05-14 National University Corporation Okayama University Pharmaceutical composition for treatment or prevention of neurodegenerative diseases
US10588881B2 (en) 2015-10-31 2020-03-17 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US11065219B2 (en) 2015-10-31 2021-07-20 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10485778B2 (en) 2015-10-31 2019-11-26 lo Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10278932B2 (en) 2015-10-31 2019-05-07 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10695312B2 (en) 2015-10-31 2020-06-30 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10702489B2 (en) 2015-10-31 2020-07-07 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10806713B2 (en) 2015-10-31 2020-10-20 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10973791B2 (en) 2015-10-31 2021-04-13 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10842764B2 (en) 2015-10-31 2020-11-24 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10857117B2 (en) 2015-10-31 2020-12-08 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10980760B2 (en) 2015-10-31 2021-04-20 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10980759B2 (en) 2015-10-31 2021-04-20 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10980761B2 (en) 2015-10-31 2021-04-20 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US10092535B2 (en) 2015-10-31 2018-10-09 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
US9877941B2 (en) 2015-10-31 2018-01-30 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of RXR agonists and thyroid hormones
KR102222619B1 (ko) * 2016-03-10 2021-03-05 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
JP7169646B2 (ja) 2016-03-10 2022-11-11 アイオー セラピューティクス インコーポレイテッド Rxrアゴニストおよび甲状腺ホルモンの組み合わせでの自己免疫疾患の治療
US10946001B2 (en) 2016-03-10 2021-03-16 Io Therapeutics, Inc. Treatment of autoimmune diseases with combinations of RXR agonists and thyroid hormones
WO2017155577A1 (en) 2016-03-10 2017-09-14 Io Therapeutics, Inc. Treatment of autoimmune diseases with combinations of rxr agonists and thyroid hormones
KR20210024227A (ko) * 2016-03-10 2021-03-04 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
KR102605349B1 (ko) * 2016-03-10 2023-11-22 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
US10835507B2 (en) 2016-03-10 2020-11-17 Io Therapeutics, Inc. Treatment of muscular disorders with combinations of RXR agonists and thyroid hormones
KR20180120754A (ko) * 2016-03-10 2018-11-06 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
IL261669B1 (en) * 2016-03-10 2023-08-01 Io Therapeutics Inc A combination of thyroid hormones and a rxr agonist for the treatment of autoimmune diseases
EP3426302A4 (en) * 2016-03-10 2019-10-30 IO Therapeutics, Inc. TREATMENT OF AUTOIMMUNE DISEASES USING COMBINATIONS OF RXR AGONISTS AND THYROID HORMONES
KR20220025918A (ko) * 2016-03-10 2022-03-03 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
US11690831B2 (en) 2016-03-10 2023-07-04 Io Therapeutics, Inc. Treatment of autoimmune diseases with combinations of RXR agonists and thyroid hormones
JP7169647B2 (ja) 2016-03-10 2022-11-11 アイオー セラピューティクス インコーポレイテッド Rxrアゴニストおよび甲状腺ホルモンの組み合わせでの筋肉障害の治療
US11690832B2 (en) 2016-03-10 2023-07-04 Io Therapeutics Treatment of autoimmune diseases with combinations of RXR agonists and thyroid hormones
JP2019511492A (ja) * 2016-03-10 2019-04-25 アイオー セラピューティクス インコーポレイテッド Rxrアゴニストおよび甲状腺ホルモンの組み合わせでの筋肉障害の治療
JP2019507777A (ja) * 2016-03-10 2019-03-22 アイオー セラピューティクス インコーポレイテッド Rxrアゴニストおよび甲状腺ホルモンの組み合わせでの自己免疫疾患の治療
EP4166160A1 (en) * 2016-03-10 2023-04-19 IO Therapeutics, Inc. Treatment of autoimmune diseases with combinations of rxr agonists and thyroid hormones
KR102549273B1 (ko) * 2016-03-10 2023-06-28 아이오 테라퓨틱스, 인크. Rxr 작용제 및 갑상선 호르몬의 조합을 사용한 자가면역 질환의 치료
US11517549B2 (en) 2017-09-20 2022-12-06 Io Therapeutics, Inc. Treatment of disease with esters of selective RXR agonists
US10590059B2 (en) 2017-11-17 2020-03-17 Io Therapeutics, Inc. Compounds and synthetic methods for the preparation of retinoid X receptor-specific retinoids
US10919835B2 (en) 2017-11-17 2021-02-16 Io Therapeutics, Inc. Compounds and synthetic methods for the preparation of retinoid X receptor-specific retinoids
US10966950B2 (en) 2019-06-11 2021-04-06 Io Therapeutics, Inc. Use of an RXR agonist in treating HER2+ cancers
US11224583B2 (en) 2019-06-11 2022-01-18 Io Therapeutics, Inc. Use of an RXR agonist in treating HER2+ cancers
US11896558B2 (en) 2021-12-07 2024-02-13 Io Therapeutics, Inc. Use of an RXR agonist and taxanes in treating Her2+ cancers

Also Published As

Publication number Publication date
WO2013090616A1 (en) 2013-06-20
AU2017210644A1 (en) 2017-10-05
US11793781B2 (en) 2023-10-24
MX352727B (es) 2017-12-06
CN110151745A (zh) 2019-08-23
AU2012352149B2 (en) 2017-06-01
US20190125705A1 (en) 2019-05-02
US10201512B2 (en) 2019-02-12
CA2858882C (en) 2021-02-02
US20210128503A1 (en) 2021-05-06
US10285960B2 (en) 2019-05-14
US20150342917A1 (en) 2015-12-03
US11166927B2 (en) 2021-11-09
AU2017210644B2 (en) 2019-04-04
US11547684B2 (en) 2023-01-10
CN104114171A (zh) 2014-10-22
US11576881B2 (en) 2023-02-14
US10695307B2 (en) 2020-06-30
MX2014007169A (es) 2016-03-04
US20210128504A1 (en) 2021-05-06
AU2012352149A1 (en) 2014-07-03
CA2858882A1 (en) 2013-06-20
US20180116985A1 (en) 2018-05-03
US20150196517A1 (en) 2015-07-16
US20200155488A1 (en) 2020-05-21
US10034845B2 (en) 2018-07-31
US20200155489A1 (en) 2020-05-21
US10945976B2 (en) 2021-03-16
US11246845B2 (en) 2022-02-15
EP2790698A1 (en) 2014-10-22
US20180263939A1 (en) 2018-09-20
US20170056348A1 (en) 2017-03-02
US20220117922A1 (en) 2022-04-21
US20220151964A1 (en) 2022-05-19

Similar Documents

Publication Publication Date Title
US11793781B2 (en) Autoimmune disorder treatment using RXR agonists
US20220370386A1 (en) Treatment of diseases by concurrently eliciting remyelination effects and immunomodulatory effects using selective rxr agonists
US11690832B2 (en) Treatment of autoimmune diseases with combinations of RXR agonists and thyroid hormones
US20120238623A1 (en) Inflammation and Autoimmune Disorder Treatment using RARa Selective Agonists
US20140194517A1 (en) Treatment of Graft-Versus-Host Disease Disorders using RAR Antagonists

Legal Events

Date Code Title Description
AS Assignment

Owner name: DARTMOUTH COLLEGE, NEW HAMPSHIRE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DMITROVSKY, ETHAN;NOWAK, ELIZABETH;NOELLE, RANDOLPH J.;SIGNING DATES FROM 20120315 TO 20121003;REEL/FRAME:029472/0109

Owner name: IO THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHANDRARATNA, ROSHANTHA A.;REEL/FRAME:029471/0884

Effective date: 20120313

AS Assignment

Owner name: IO THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHANDRARATNA, ROSHANTHA A.;REEL/FRAME:029587/0332

Effective date: 20121219

AS Assignment

Owner name: TRUSTEES OF DARTMOUTH COLLEGE, NEW HAMPSHIRE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DMITROVSKY, ETHAN;NOWAK, ELIZABETH;NOELLE, RANDOLPH;SIGNING DATES FROM 20121219 TO 20130124;REEL/FRAME:029706/0756

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:DARTMOUTH COLLEGE;REEL/FRAME:041420/0330

Effective date: 20170113

STCB Information on status: application discontinuation

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