WO2020106741A1 - Compounds and compositions for treating conditions associated with sting activity - Google Patents

Compounds and compositions for treating conditions associated with sting activity

Info

Publication number
WO2020106741A1
WO2020106741A1 PCT/US2019/062245 US2019062245W WO2020106741A1 WO 2020106741 A1 WO2020106741 A1 WO 2020106741A1 US 2019062245 W US2019062245 W US 2019062245W WO 2020106741 A1 WO2020106741 A1 WO 2020106741A1
Authority
WO
WIPO (PCT)
Prior art keywords
independently selected
optionally substituted
ring
cancer
alkyl
Prior art date
Application number
PCT/US2019/062245
Other languages
French (fr)
Inventor
William R. Roush
Hans Martin Seidel
Shankar Venkatraman
Original Assignee
Ifm Due, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ifm Due, Inc. filed Critical Ifm Due, Inc.
Publication of WO2020106741A1 publication Critical patent/WO2020106741A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4402Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 2, e.g. pheniramine, bisacodyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/453Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes (“STING”) or inhibit cyclic GMP-AMP Synthase (“cGAS”).
  • chemical entities e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound
  • STING Stimulator of Interferon Genes
  • cGAS cyclic GMP-AMP Synthase
  • Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human).
  • This disclosure also features compositions containing the same as well as methods of using and making the same.
  • the cGAS/STING pathway is a component of inflammatory signaling pathways.
  • cGAMP cyclic GMP-AMP
  • STING induces the phosphorylation of and nuclear translocation of interferon (IFN) regulatory factors (IRFs).
  • IFN interferon regulatory factors
  • IRFs regulate the expression of genes, including the type I IFNs, which regulate the activity of the immune system.
  • STING also known as transmembrane protein 173 (TMEM173) and
  • MPYS/MITA/ERIS is a protein that in humans is encoded by the TMEM173 gene.
  • STING has been shown to play a role in innate immunity. STING induces type I interferon production when cells are infected with intracellular pathogens, such as viruses, mycobacteria and intracellular parasites. Type I interferon, mediated by STING, protects infected cells and nearby cells from local infection in an autocrine and paracrine manner.
  • STING a transmembrane protein localized to the endoplasmic reticulum (ER) acts as a second messenger receptor for 2', 3' cyclic GMP-AMP (hereafter cGAMP), which is produced by cGAS after dsDNA binding.
  • cGAMP 2', 3' cyclic GMP-AMP
  • STING can also function as a primary pattern recognition receptor for bacterial cyclic dinucleotides (CDNs) and small molecule agonists.
  • CDNs bacterial cyclic dinucleotides
  • Ligand-induced activation of STING triggers its re-localization to the Golgi, a process essential to promote the interaction of STING with TBK1.
  • This protein complex signals through the transcription factors IRF-3 to induce type I interferons (IFNs) and other co-regulated antiviral factors.
  • IFNs type I interferons
  • STING was shown to trigger NF-KB and MAP kinase activation. Following the initiation of signal transduction, STING is rapidly degraded, a step considered important in terminating the inflammatory response.
  • STING-associated vasculopathy with onset in infancy SAVI
  • TMEM173 the gene name of STING
  • STING is implicated in the pathogenesis of Aicardi- Goutieres Syndrome (AGS) and genetic forms of lupus.
  • AGS Aicardi- Goutieres Syndrome
  • This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes (“STING”) or inhibit cyclic GMP-AMP Synthase (“cGAS”).
  • chemical entities e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound
  • STING Stimulator of Interferon Genes
  • cGAS cyclic GMP-AMP Synthase
  • Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human).
  • This disclosure also features compositions containing the same as well as methods of using and making the same.
  • the term“STING antagonist” is an agent that decreases one or both of (i) the activity of STING (e.g., any of the exemplary activities of STING described herein) (e.g., as compared to the level of STING activity in the absence of the agent) and (ii) the expression level of STING in a mammalian cell (e.g., using any of the exemplary methods of detection described herein) (e.g., as compared to the expression level of STING in a mammalian cell not contacted with the agent) .
  • An "antagonist” of STING also includes compounds that, at the protein level, directly bind or modify STING such that an activity of STING is decreased, e.g., by inhibition, blocking or dampening agonist-mediated responses, altered distribution, or otherwise.
  • STING antagonists include chemical entities, which interfere or inhibit STING signaling.
  • cGAS inhibitor is an agent that decreases one or both of (i) the activity of cGAS (e.g., any of the exemplary activities of cGAS described herein) (e.g., as compared to the level of cGAS activity in the absence of the agent) and (ii) the expression level of cGAS in a mammalian cell (e.g., using any of the exemplary methods of detection described herein) (e.g., as compared to the expression level of cGAS in a mammalian cell not contacted with the agent).
  • the activity of cGAS e.g., any of the exemplary activities of cGAS described herein
  • the expression level of cGAS in a mammalian cell e.g., using any of the exemplary methods of detection described herein
  • an “inhibitor” of cGAS also includes compounds that, at the protein level, directly bind or modify cGAS such that an activity of cGAS is decreased, e.g., by inhibition, blocking or dampening agonist-mediated responses, altered distribution, or otherwise.
  • cGAS inhibitors include chemical entities, which interfere or inhibit cGAS signaling.
  • R 1 , R 2 , R 3 , R 4 , R 5 , W, Q, and A can be as defined anywhere herein.
  • compositions are featured that include a chemical entity described herein (e.g., a formula (I) compound described genetically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) and one or more pharmaceutically acceptable excipients.
  • a chemical entity described herein e.g., a formula (I) compound described genetically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same
  • one or more pharmaceutically acceptable excipients are featured that include a chemical entity described herein (e.g., a formula (I) compound described genetically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) and one or more pharmaceutically acceptable excipients.
  • methods for inhibiting the STING pathway e.g., inhibiting (e.g., antagonizing) STING activity or inhibiting cGAS activity are featured that include contacting STING or cGAS with a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
  • Methods include in vitro methods, e.g., contacting a sample that includes one or more cells comprising STING or cGAS (e.g., innate immune cells, e.g., mast cells, macrophages, dendritic cells (DCs), and natural killer cells) with the chemical entity.
  • a chemical entity described herein e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same.
  • Methods include in vitro methods, e.g., contacting a sample that includes one or more cells comprising STING or c
  • Methods can also include in vivo methods; e.g., administering the chemical entity to a subject (e.g., a human) having a disease in which increased (e.g., excessive) STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the disease.
  • a subject e.g., a human
  • increased (e.g., excessive) STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the disease.
  • methods of treating a condition, disease or disorder ameliorated by antagonizing STING or inhibiting cGAS are featured, e.g., treating a condition, disease or disorder in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human).
  • the methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
  • methods of treating cancer include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
  • a chemical entity described herein e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same.
  • methods of treating other STING or cGAS-associated conditions are featured, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.
  • the methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
  • methods of suppressing STING-dependent type I interferon production in a subject in need thereof include administering to the subject an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
  • a chemical entity described herein e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same.
  • methods of treating a disease in which increased (e.g., excessive) STING or cGAS activation e.g., STING or cGAS signaling
  • the methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
  • methods of treatment include administering an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) to a subject; wherein the subject has (or is predisposed to have) a disease in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the disease .
  • a chemical entity described herein e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same
  • STING or cGAS activation e.g., STING or cGAS signaling
  • methods of treatment that include administering to a subject a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same), wherein the chemical entity is administered in an amount effective to treat a disease in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.
  • a chemical entity described herein e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same
  • STING or cGAS activation e.g., STING or cGAS signaling
  • Embodiments can include one or more of the following features.
  • the chemical entity can be administered in combination with one or more additional therapeutic agents and/or regimens.
  • methods can further include administering one or more (e.g., two, three, four, five, six, or more) additional agents.
  • the chemical entity can be administered in combination with one or more additional therapeutic agents and/or regimens that are useful for treating other STING or cGAS-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.
  • type I interferonopathies e.g., STING-associated vasculopathywith onset in infancy (SAVI)
  • Aicardi-Goutieres Syndrome (AGS) e.g., STING-associated vasculopathywith onset in infancy (SAVI)
  • Aicardi-Goutieres Syndrome (AGS) e.g., Aicardi-Goutieres Syndrome (AGS)
  • genetic forms of lupus e.g., STING
  • the chemical entity can be administered in combination with one or more additional cancer therapies (e.g., surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof; e.g., chemotherapy that includes administering one or more (e.g., two, three, four, five, six, or more) additional chemotherapeutic agents.
  • additional cancer therapies e.g., surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof; e.g., chemotherapy that includes administering one or more (e.g., two, three, four, five, six, or more) additional chemotherapeutic agents.
  • Non-limiting examples of additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g.,azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisom erase; e.g., camptothecins, such as irinotecan and/or topotecan;.
  • an alkylating agent e.g.,
  • LAG3 MHC class II - LAG3, 4-1BB-4-1BB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM
  • HVEM-BTL A-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48
  • CD244 CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2,
  • HHLA2-TMIGD2 Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PDl or PD-L1).
  • CTLA-4 or PDl or PD-L1 CTLA-4 or PDl or PD-L1
  • the subject can have cancer; e.g., the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies.
  • Non-limiting examples of cancer include melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.
  • the cancer can be a refractory cancer.
  • the chemical entity can be administered intratumorally.
  • the methods can further include identifying the subject.
  • STING is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous STING molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.
  • cGAS is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous cGAS molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.
  • API refers to an active pharmaceutical ingredient.
  • an“effective amount” or“therapeutically effective amount,” as used herein, refer to a sufficient amount of a chemical entity (e.g., a formula (I) compound or a pharmaceutically acceptable salt and/or hydrate and/or cocrystal thereof) being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated.
  • the result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an“effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms.
  • An appropriate“effective” amount in any individual case is determined using any suitable technique, such as a dose escalation study.
  • excipient or “pharmaceutically acceptable excipient” means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material.
  • each component is“pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-m ethy 1 -D-gl ucam i n e, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
  • a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-m ethy 1 -D-gl ucam i n e, tris(hydroxymethyl)methylamine, and
  • Examples of a salt that the compounds described hereinform with a base include the following: salts thereof with inorganic bases such as sodium, potassium, magnesium, calcium, and aluminum; salts thereof with organic bases such as methylamine, ethylamine and ethanolamine; salts thereof with basic amino acids such as lysine and ornithine; and ammonium salt.
  • the salts may be acid addition salts, which are specifically exemplified by acid addition salts with the following: mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid:organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; acidic amino acids such as aspartic acid and glutamic acid.
  • mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid
  • organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tart
  • composition refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents.
  • excipients such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.
  • subject refers to an animal, including, but not limited to, a primate (e.g ., human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse.
  • primate e.g ., human
  • monkey cow, pig, sheep, goat
  • horse dog, cat, rabbit, rat
  • patient refers to a mammalian subject, such as a human.
  • treat in the context of treating a disease or disorder, are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof.
  • The“treatment of cancer” refers to one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, (i) slowing down and (ii) complete growth arrest; (2) reduction in the number of tumor cells; (3) maintaining tumor size; (4) reduction in tumor size; (5) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of tumor cell infiltration into peripheral organs; (6) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of metastasis; (7) enhancement of anti-tumor immune response, which may result in (i) maintaining tumor size, (ii) reducing tumor size, (iii) slowing the growth of a tumor, (iv) reducing, slowing or preventing invasion and/or (8) relief, to some extent, of the severity or number of one or more symptoms associated with the disorder.
  • halo refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I).
  • alkyl refers to a hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms.
  • Ci-io indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it.
  • Non-limiting examples include methyl, ethyl, No-propyl, N/V-butyl, «-hexyl.
  • haloalkyl refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo.
  • alkoxy refers to an -O-alkyl radical (e.g., -OCFE).
  • alkylene refers to a divalent alkyl (e.g., -CFh-).
  • alkenyl refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds.
  • the alkenyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it.
  • alkynyl refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds.
  • the alkynyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it.
  • aryl refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent.
  • aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like.
  • cycloalkyl as used herein includes cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted.
  • cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Cycloalkyl may include multiple fused and/or bridged rings.
  • Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butane, bicyclo[2.1.0]pentane, bicyclo[l . l . l]pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like.
  • Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • spirocyclic cycloalkyls include spiro[2.2]pentane, spiro[2.5]octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro[4.5]decane, spiro[3.6]decane, spiro[5.5]undecane, and the like.
  • cycloalkenyl as used herein includes partially unsaturated cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkenyl group may be optionally substituted.
  • Examples of cycloalkenyl groups include, without limitation, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl.
  • Cycloalkenyl groups may have any degree of saturation provided that none of the rings in the ring system are aromatic; and the cycloalkenyl group is not fully saturated overall. Cycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.
  • heteroaryl means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl), and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S.
  • heteroatoms independently selected from the group consisting of N, O, and S.
  • Heteroaryl groups can either be unsubstituted or substituted with one or more substituents.
  • heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3- ⁇ i]pyr
  • the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl.
  • heterocyclyl refers to a mon-, bi-, tri-, or polycyclic nonaromatic ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent.
  • ring atoms e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system
  • heteroatoms selected from O, N, or S (e
  • heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like.
  • Heterocyclyl may include multiple fused and bridged rings.
  • fused/bridged heteorocyclyl includes: 2-azabicyclo[1.1.0]butane, 2-azabicyclo[2.1.0]pentane, 2- azabicyclo[l. l.
  • Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • spirocyclic heterocyclyls include 2- azaspiro[2.2]pentane, 4-azaspiro[2.5]octane, l-azaspiro[3.5]nonane, 2- azaspiro[3.5]nonane, 7-azaspiro[3.5]nonane, 2-azaspiro[4.4]nonane, 6- azaspiro[2.6]nonane, l,7-diazaspiro[4.5]decane, 7-azaspiro[4.5]decane 2,5- diazaspiro[3 6]decane, 3-azaspiro[5.5]undecane, 2-oxaspiro[2.2]pentane, 4- oxaspiro[2.5]octane, l-oxaspiro
  • atoms making up the compounds of the present embodiments are intended to include all isotopic forms of such atoms.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium
  • isotopes of carbon include 13 C and 14 C.
  • This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes (“STING”) or inhibit cyclic GMP-AMP Synthase (“cGAS”).
  • chemical entities e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound
  • STING Stimulator of Interferon Genes
  • cGAS cyclic GMP-AMP Synthase
  • Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human).
  • This disclosure also features compositions containing the same as well as methods of using and making the same.
  • W is selected from the group consisting of:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • Q-A is defined according to (A) or (B) below: ⁇
  • Q is NH or N(R q ), wherein R q is Ci-6 alkyl which is optionally substituted with from 1-2 independently selected R a ; or
  • R q and R 4 taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each of the heteroatoms is independently selected from N, N(H), O, and S(0)o-2.
  • A is:
  • n 0 or 1
  • Ci-6 alkylene which is optionally substituted with from 1-6 R a and further optionally substituted with one oxo;
  • heteroaryl including from 5-20 ring atoms, wherein from 1- 4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c , or
  • heterocyclyl including from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected R b , OR
  • Ci-io alkyl which is optionally substituted with from 1-6 independently selected
  • E is heterocyclyl including from 3-16 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected R b ;
  • R 1 is selected from the group consisting of:
  • R 2 is selected from the group consisting of:
  • Ci-6 alkyl optionally substituted with 1-2 R a , and heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c ; or R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci-3 alkyl, halo, hydroxy, and
  • Ci-6 alkyl which is optionally substituted with from 1-2 R a ,
  • Ci-6 alkoxy which is optionally substituted with from 1-2 R a ,
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c ,
  • R 3 and R 4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci- 3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2;
  • each of R 4A , R 4B , R 5A and R 5B is independently selected from the group consisting of:
  • W 1 is Ci-3 alkylene, which is optionally substituted with from 1-6 R a ;
  • heteroaryl including from 5-10 ring atoms, wherein from 1- 4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c ; or
  • heterocyclyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected R b ; each occurrence of R 6A is independently:
  • Ci-10 alkyl which is optionally substituted with 1-6 independently selected R a ;
  • (vi) (Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected R b ; or
  • Ci-io alkyl which is optionally substituted with from 1-6 independently selected
  • heterocyclyl includes from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclyl is optionally substituted with from 1-4 independently selected Ci-4 alkyl;
  • R d is selected from the group consisting of: Ci-6 alkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci- 4 alkyl); -CON(R’)(R”); -S(0)I-2(NR’R”); - S(0)i- 2 (Ci-4 alkyl); -OH; and Ci-4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; Ci- 6 alkyl; Ci- 6 haloalkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); - CON(R’)(R”); -S(0)I- 2 (NR’R”); - S(0)i- 2 (Ci-4 alkyl); -OH; and CM alkoxy; or R e and R f together with the nitrogen atom to which each is attached forms a ring including from 3-8
  • W is selected from the group consisting of:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g.,
  • Q-A is defined according to (A) or (B) below:
  • Q is NH or N(R q ), wherein R q is Ci-6 alkyl which is optionally substituted with from 1-2 independently selected R a ; or
  • R q and R 4 taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each of the heteroatoms is independently selected from N, N(H), O, and S(0)o-2.
  • A is:
  • n 0 or 1
  • Ci-6 alkylene which is optionally substituted with from 1-6 R a ;
  • heterocyclyl including from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1- 4 independently selected R b ,
  • Ci-io alkyl which is optionally substituted with from 1-6 independently selected R a , or
  • E is heterocyclyl including from 3-16 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected R b ;
  • R 1 is selected from the group consisting of:
  • R 2 is selected from the group consisting of: H, halo, cyano, 0C(0)R 4B , NHC(0)R 4B , OR 5B , SR 5B , NHS0 2 R 4B , 0P(0)(0R 5B ) 2 , Ci-e alkyl optionally substituted with 1-2 R a , and heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c ; or
  • R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci- 3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 ; each of R 3 , R 4 , and R 5 is independently selected from the group consisting of:
  • Ci-6 alkyl which is optionally substituted with from 1-2 R a ,
  • Ci-6 alkoxy which is optionally substituted with from 1-2 R a ,
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c ,
  • R 3 and R 4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 ; each of R 4A , R 4B , R 5A and R 5B is independently selected from the group consisting of:
  • Ci-6 alkyl optionally substituted with 1-6 R a ;
  • W 1 is Ci-3 alkylene, which is optionally substituted with from 1-6 R a ;
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c ; or
  • heterocyclyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2 and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1- 4 independently selected R b ; each occurrence of R 6A is independently:
  • Ci-10 alkyl which is optionally substituted with 1-6 independently selected R a ;
  • Ci-4 alkoxy or two occurrences of R 6A together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R 6 ), which are each independently selected from the group consisting of N(H), N(R d ), O, and S(0)o-2; each occurrence of R a is independently selected from the group consisting of: -
  • Ci-10 alkyl which is optionally substituted with from 1-6 independently selected R a ;
  • C2-6 alkenyl
  • heterocyclyl includes from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein the heterocyclyl is optionally substituted with from 1-4 independently selected Ci-4 alkyl;
  • R d is selected from the group consisting of: Ci-6 alkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); -CON(R’)(R”); -S(0)I-2(NR’R”); - S(0)i- 2 (Ci-4 alkyl); -OH; and Ci- 4 alkoxy; each occurrence of R e and R f is independently selected from the group consisting of: H; Ci-6 alkyl; Ci-6 haloalkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); - CON(R’)(R”); -S(0)I-2(NR , R”); - S(0)i- 2 (Ci-4 alkyl); -OH; and CM alkoxy; or R e and R f together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring
  • Embodiments can include any one or more of the features delineated below and/or in the claims.
  • R 1 is selected from NO2, S02R 4A , and S(0)2N(R 6A )2.
  • R 1 is S02R 4A .
  • R 4A is Ci- 6 alkyl optionally substituted with 1- 6 R a ; or (iii) -(W 4 ) q -W 2 )
  • R 4A is Ci- 6 alkyl optionally substituted with 1-3 R a .
  • R 4A is unsubstituted Ci- 6 alkyl (e.g., C 1-3 alkyl, e.g., methyl).
  • R 1 can be S(0)2Me.
  • R 4A is Ci- 6 alkyl optionally substituted with 1-3 R a
  • R 4A is Ci- 6 alkyl substituted with from 1-3 R a (e.g., trifluoromethyl).
  • R 1 can be S(0)2CF3.
  • R 4A is -(W 4 ) q -W 2 . In certain embodiments, q is 0. In other embodiments, q is 1.
  • W 1 is C 1-3 alkylene (e.g., CH2).
  • W 2 is C6-10 aryl, which is optionally substituted with from
  • W 2 is phenyl, which is optionally substituted with from 1-2 R c (e.g., W 2 is unsubstituted phenyl).
  • R 1 can be S(0)2Ph.
  • W 2 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c .
  • W 2 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c .
  • W 2 is heteroaryl including from 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c .
  • W 2 can be pyridinyl optionally substituted with from 1-2 independently selected R c (e.g., W 2 can be pyridin-4-yl or pyridine-2 -yl, e.g., W 2 can be unsubstituted pyridin-4-yl or pyridin-2-yl).
  • W 2 is heteroaryl including from 5 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c .
  • W 2 can be thiazolyl optionally substituted with from 1-2 independently selected R c (e.g., W 2 can be thiazol-5-yl optionally substituted with 1 R c ).
  • R 1 can be selected from:
  • R 1 is NO2.
  • R 1 is S(0)2N(R 6A )2. In certain of these embodiments, R 1 is S(0)2NHR 6A . In certain of these embodiments, R 6A is H or Ci-6 alkyl which is optionally substituted with from 1-6 R 6 (e.g., H or unsubstituted Ci-6 alkyl). As non-limiting examples, R 6A can be S(0)2NH2, S(0)2NHMe, or S ⁇ NffBu.
  • R 1 is C(0)N(R 6A )2.
  • each of R 6A is independently selected from:
  • Ci-6 alkyl optionally substituted with from 1-6 R a .
  • one R 6A is H; and the other R 6A is Ci-6 alkyl optionally substituted with from 1-6 R a .
  • R 1 can be S(0)2NHMe; or R 1 can be C(0)NHMe.
  • R 1 is CN
  • R 1 is F.
  • R 5A is selected from (i) H; and (ii) Ci- 6 alkyl optionally substituted with from 1-6 R a (e.g., unsubstituted Ci- 6 alkyl, e.g., ethyl or methyl).
  • R 4A is H.
  • R 2 is selected from the group consisting of: H, halo, cyano, 0C(0)R 4B , NHC(0)R 4B , OR 5B , and SR 5B
  • R 2 is selected from the group consisting of: H, halo, cyano, 0C(0)R 4B , and SR 5B
  • R 2 is selected from H, halo, and cyano.
  • R 2 is H.
  • R 2 is halo
  • R 2 is cyano.
  • R 4 is F or C(0)N(R 6A )2 (e.g., R 1 is F).
  • each of R 4B and R 5B is independently selected from: (ii) Ci- 6 alkyl optionally substituted with 1-6 R a ; and (iii) -(W 4 ) q -W 2 (e.g., q is 0; or q is 1).
  • each of R 4B and R 5B is independently (iii) - (W 4 ) q -W 2 (e.g., q is 0; or q is 1). In certain embodiments of foregoing, q is 0.
  • W 2 is selected from:
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c .
  • W 2 can be phenyl, which is optionally
  • W 2 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected R c .
  • W 2 is selected heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c .
  • W 2 can
  • Ci-6 alkyl is unsubstituted Ci-6 alkyl
  • R 5B is heteroaryl including from 5- 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently
  • R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes:
  • R 2 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c , such as R 2 is imidazolyl optionally substituted with one R c .
  • R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2.
  • R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring is substituted with from 1-2 oxo.
  • R 1 and R 2 taken together with the atoms to which each is attached can form a ring selected from the following:
  • R 1 is S02R 4A or S(0)I-2N(R 6A )2; and R 2 is halo such as chloro.
  • R 4A is Ci- 6 alkyl; and each R 6A is independently Ci-io alkyl or H.
  • R 1 can be S(0)2Me or S(0)2NHMe; and R 2 can be chloro.
  • R 4A is Ci- 6 alkyl.
  • each of R 3 , R 4 , and R 5 is independently selected from the group consisting of:
  • Ci- 6 alkyl which is optionally substituted with from 1-2 R a ,
  • Ci- 6 alkoxy which is optionally substituted with from 1-2 R a , and
  • Ci-6 haloalkoxy which is optionally substituted with from 1-2 R a .
  • each of R 3 , R 4 , and R 5 is independently selected from the group consisting of:
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci-6 alkyl e.g., methyl
  • R 3 is H.
  • R 3 is halo (e.g., F).
  • R 3 is Ci-6 alkyl (e.g., methyl).
  • R 4 is selected from the group consisting of:
  • Ci-6 alkoxy e.g., methoxy
  • R 4 is H.
  • R 5 is H.
  • R 3 and R 4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 .
  • R 3 and R 4 taken together with the carbon to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 .
  • R 3 and R 4 taken together with the carbon to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 .
  • R 3 and R 4 taken together with the carbon to which each is attached can form a ring selected from the following:
  • W is selected from:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • W is C1-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g., F).
  • W is unsubstituted C1-3 alkylene (e.g., CFh).
  • W is C1-3 alkylene substituted with from 1-4 fluoro (e.g., -C(H)(CF 3 )-).
  • Q and A are defined according to (A).
  • Q is NFL
  • A is -(Y A1 ) n -Y A2 . In certain embodiments, n is 0. In some embodiments, n is 1.
  • Y A1 is C1-3 alkylene (e.g., Y A1 is CFh or CH2CH2).
  • Y A2 is C6-20 aryl, which is optionally substituted with from
  • Y A2 is C6-10 aryl, which is optionally substituted with from
  • Y A2 is phenyl, which is optionally substituted with from
  • Y A2 is phenyl which is substituted with 1 R c .
  • Y A2 can be phenyl substituted with R c at the para position.
  • Y A2 is C9-10 aryl, which is optionally substituted with from 1-2 R c .
  • Y A2 can be tetrahydro-naphthyl, which
  • Y A2 is optionally substituted with from 1-3 R c (e.g., Y A2 can be
  • Y A2 is heteroaryl including from 6-10 (e.g., 8-10) ring atoms, wherein from 1-4 (e.g., from 1-2, e.g., 1) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c .
  • Y A2 heteroaryl including from 8-10 ring atoms, wherein from 1-2, such as 1, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c .
  • Y ⁇ can be
  • Y A2 is heteroaryl including 5-6, such as 6 ring atoms, wherein from 1-2, such as 1, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected R c .
  • Y A2 is pyridyl which is optionally substituted with from 1-2 independently selected R c .
  • Y ⁇ 2 can be: .
  • each occurrence of R c is independently selected from Ci-io alkyl which is optionally substituted with 1-2 independently selected R a , Ci-4 alkoxy, and C3-6 cycloalkyl.
  • each R a substituent of R c is independently selected from hydroxy and Ci- 6 alkoxy.
  • each occurrence of R c is independently selected from butyl (e.g., n-butyl), propyl (e.g., n-propyl), cyclohexyl, 2-hydroxybut-l-yl, ethoxymethyl, methoxy, and ethoxy.
  • each occurrence of R c is independently C3-6 cycloalkyl such as cyclobutyl and cyclohexyl or C6-10 aryl such as phenyl.
  • A is e.g., R c is as defined supra or in claims
  • R c is n-butyl, cyclohexyl, 2-hydroxybut-l-yl, or ethoxymethyl).
  • a A i iss ⁇ — " ( (ee.g g. , , ) (e.g ⁇ , R c is as defined supra or in claims 75-77, e.g., R c is ethoxy or propyl).
  • A is R c is as defined supra or in claims 75-77, e.g., R c is ethoxy).
  • Y A2 is C3-20 cycloalkyl, which is optionally substituted with from 1-4 R b .
  • Y A2 is C5-10 cycloalkyl, which is optionally substituted with from 1-4 R b .
  • Y A2 is C6-8 cycloalkyl, which is optionally substituted with from 1-2 R b .
  • each occurrence of R b is selected from Ci-10 alkyl optionally substituted with from 1-6 independently selected R a ; C i-4 haloalkyl; -OH; oxo; -F; -Cl; - Br; Ci-4 alkoxy; Ci-4 haloalkoxy; and C3-6 cycloalkyl optionally substituted with from 1-4 independently selected C i-4 alkyl.
  • each occurrence of R b is Ci-10 alkyl optionally substituted with from 1-6 independently selected R a ; Ci-4 haloalkyl; -F; -Cl; -Br; and Ci-4 alkoxy.
  • each occurrence of R b is Ci- 6 alkyl optionally substituted with from 1-3 independently selected R a ; -F; and Ci-4 alkoxy.
  • each occurrence of R b is n-propyl; n-butyl; ethoxymethyl; -F; and Ci-4 alkoxy (e.g., -F, n-propyl, and ethoxymethyl).
  • each occurrence of R b is C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl or C6-10 aryl.
  • A is selected from:
  • R b is as defined in claims 84-87 (e.g., R b is F; or R b is n-propyl; or R b is ethoxymethyl).
  • Q is N(R q ).
  • R q and R 4 taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each heteroatom is independently selected from N, N(H), O, and S(0)o-2.
  • R q and R 4 taken together with the atoms connecting them, forms a ring including 5-6 ring atoms, wherein the ring includes (a) from 2-5 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each heteroatom is independently selected from N, N(H), O, and S(0)o-2.
  • Q and A are defined according to (B).
  • E is heterocyclyl including from 3-12 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2 and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-2 independently selected R b .
  • E is heterocyclyl including from 3-12 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2,and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1 independently selected R b .
  • E is: L
  • the compound is of Formula (1-1):
  • R cl is as defined for R c .
  • R cl is as defined for R c ; and wherein said R c is as defined in any one of claims 75-77.
  • R cl is selected from Ci-io alkyl which is optionally substituted with 1-2 independently selected R a , Ci-4 alkoxy, and C3-6 cycloalkyl.
  • R cl is C3-6 cycloalkyl such as cyclobutyl and cyclohexyl or C6-10 aryl such as phenyl.
  • each each R a substituent of R c is independently selected from hydroxy and Ci-6 alkoxy.
  • R c can be selected from butyl (e.g., n-butyl), propyl (e.g., n-propyl), cyclohexyl, 2-hydroxybut-l-yl, ethoxymethyl, methoxy, and ethoxy.
  • R c can be CF3.
  • W is selected from:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • R 1 is as defined in any one of claims 2-29 (e.g., R 1 is NO2).
  • R 1 is SCbR 4A .
  • R 4A is Ci- 6 alkyl optionally substituted with 1-6 R a (e.g., R 4A is unsubstituted Ci- 6 alkyl such as methyl).
  • R 1 can be S(0)2Me.
  • R 1 can be S(0)2N(R 6A )2.
  • each R 6A is independently H or Ci-10 alkyl.
  • R 1 can be S(0) 2 NHMe.
  • R 2 is as defined in any one of claims 30-41.
  • R 2 is selected from the group consisting of: H, halo, cyano, OC(0)R 4B , and SR 5B (e.g., R 2 can be selected from H, halo, and cyano).
  • R 2 is halo (e.g., chloro).
  • R 2 is halo; and R 1 is SCbR 4A or S(0)2N(R 6A )2.
  • R 2 can be chloro; and R 1 can be SCbR 4A or S(0) 2 N(R 6A )2, wherein R 4A is Ci- 6 alkyl; and each R 6A is independently H or Ci-10 alkyl.
  • R 1 and R 2 are as defined in any one of claims 42-45.
  • each of R 3 , R 4 , and R 5 is independently selected from the group consisting of:
  • Ci- 6 alkyl which is optionally substituted with from 1-2 R a
  • Ci-6 alkoxy which is optionally substituted with from 1-2 R a
  • Ci-6 haloalkoxy which is optionally substituted with from 1-2 R a .
  • each of R 3 , R 4 , and R 5 is independently selected from the group consisting of:
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci-6 alkyl e.g., methyl
  • R 3 is H.
  • R 4 is selected from the group consisting of:
  • Ci-6 alkoxy e.g., methoxy
  • R 4 is H.
  • each of R 3 and R 4 is H.
  • R 3 and R 4 is as defined in any one of claims 51-54.
  • the compound is of Formula (1-2):
  • R c2 is as defined for R c .
  • R c2 is as defined for R c ; and wherein said R c is as defined in any one of claims 75-77.
  • W is selected from:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • R 1 is as defined in any one of claims 2-29 (e.g., R 1 is NO2).
  • R 1 is S02R 4A .
  • R 4A is Ci-6 alkyl optionally substituted with 1-6 R a (e.g., R 4A is unsubstituted Ci-6 alkyl such as methyl).
  • R 1 can be S(0)2Me.
  • R 1 can be S(0)2N(R 6A )2.
  • each R 6A is independently H or Ci-10 alkyl.
  • R 1 can be S(0) 2 NHMe.
  • R 2 is as defined in any one of claims 30-41.
  • R 2 is selected from the group consisting of: H, halo, cyano, OC(0)R 4B , and SR 5B (e.g., R 2 can be selected from H, halo, and cyano).
  • R 2 is halo (e.g., chloro).
  • R 2 is halo; and R 1 is S02R 4A or S(0)2N(R 6A )2.
  • R 2 can be chloro; and R 1 can be S02R 4A or S(0)2N(R 6A )2, wherein R 4A is Ci-6 alkyl; and each R 6A is independently H or Ci-io alkyl.
  • R 1 and R 2 are as defined in any one of claims 42-45.
  • each of R 3 , R 4 , and R 5 is as defined in any one of claims 46-47.
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci-6 alkyl e.g., methyl
  • R 3 is H.
  • R 4 is selected from the group consisting of:
  • Ci-6 alkoxy e.g., methoxy
  • R 4 is H.
  • each of R 3 and R 4 is H.
  • R 3 and R 4 is as defined in any one of claims 51-54.
  • the compound is of Formula (I-3a), (I-3b), or (I-3c):
  • each R b3 is an independently selected R b .
  • each R b3 is an independently selected R b ; and wherein said R b is as defined in any one of claims 84-87.
  • W is selected from:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • R 1 is as defined in any one of claims 2-29 (e.g., R 1 is NO2).
  • R 1 is S02R 4A .
  • R 4A is Ci-6 alkyl optionally substituted with 1-6 R a (e.g., R 4A is unsubstituted Ci-6 alkyl such as methyl).
  • R 1 can be S(0)2Me.
  • R 1 can be S(0)2N(R 6A )2.
  • each R 6A is independently H or Ci-10 alkyl.
  • R 1 can be S(0) 2 NHMe.
  • R 2 is as defined in any one of claims 30-41.
  • R 2 is selected from the group consisting of: H, halo, cyano, OC(0)R 4B , and SR 5B (e.g., R 2 can be selected from H, halo, and cyano).
  • R 2 is halo (e.g., chloro).
  • R 2 is halo; and R 1 is S02R 4A or S(0)2N(R 6A )2.
  • R 2 can be chloro; and R 1 can be S02R 4A or S(0)2N(R 6A )2, wherein R 4A is Ci-6 alkyl; and each R 6A is independently H or Ci-10 alkyl.
  • R 1 and R 2 are as defined in any one of claims 42-45.
  • each of R 3 , R 4 , and R 5 is as defined in any one of claims 46-47.
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci-6 alkyl e.g., methyl
  • R 3 is H.
  • R 4 is selected from the group consisting of:
  • Ci- 6 alkoxy e.g., methoxy
  • R 4 is H.
  • each of R 3 and R 4 is H.
  • R 3 and R 4 is as defined in any one of claims 51-54.
  • the compound is of Formula (1-4)
  • R c4 is as defined for R c .
  • R c4 is as defined for R c ; and wherein said R c is as defined in any one of claims 75-77.
  • W is selected from:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • R 1 is as defined in any one of claims 2-29 (e.g., R 1 is NO2).
  • R 1 is S02R 4A .
  • R 4A is Ci-6 alkyl optionally substituted with 1-6 R a (e.g., R 4A is unsubstituted Ci-6 alkyl such as methyl).
  • R 1 can be S(0)2Me.
  • R 1 can be S(0)2N(R 6A )2.
  • each R 6A is independently H or Ci-10 alkyl.
  • R 1 can be S(0) 2 NHMe.
  • R 2 is as defined in any one of claims 30-41.
  • R 2 is selected from the group consisting of: H, halo, cyano, OC(0)R 4B , and SR 5B (e.g., R 2 can be selected from H, halo, and cyano).
  • R 2 is halo (e.g., chloro).
  • R 2 is halo; and R 1 is S02R 4A or S(0)2N(R 6A )2.
  • R 2 can be chloro; and R 1 can be S02R 4A or S(0)2N(R 6A )2, wherein R 4A is Ci-6 alkyl; and each R 6A is independently H or Ci-10 alkyl.
  • R 1 and R 2 are as defined in any one of claims 42-45.
  • each of R 3 , R 4 , and R 5 is as defined in any one of claims 46-47.
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci-6 alkyl e.g., methyl
  • R 3 is H.
  • R 4 is selected from the group consisting of:
  • R 4 is H.
  • R 5 is H.
  • each of R 3 and R 4 is H.
  • R 3 and R 4 is as defined in any one of claims 51-54.
  • the compound is of Formula (1-5)
  • R c5 is as defined for R c .
  • R c5 is as defined for R c ; and wherein said R c is as defined in any one of claims 75-77.
  • W is selected from:
  • Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
  • R 1 is as defined in any one of claims 2-29 (e.g., R 1 0 2 ).
  • R 2 is as defined in any one of claims 30-41.
  • R 1 and R 2 are as defined in any one of claims 42-45.
  • each of R 3 , and R 5 is as defined in any one of claims-47.
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci-6 alkyl e.g., methyl
  • R 5 is H.
  • each of R 3 and R 5 is H.
  • the compound is of Formula (1-6):
  • W is selected from:
  • R 1 is as defined in any one of claims 2-29 (e.g., R 1 is NO2).
  • R 1 is S02R 4A .
  • R 4A is Ci- 6 alkyl optionally substituted with 1-6 R a (e.g., R 4A is unsubstituted Ci- 6 alkyl such as methyl).
  • R 1 can be S(0)2Me.
  • R 1 can be S(0)2N(R 6A )2.
  • each R 6A is independently H or Ci-10 alkyl.
  • R 1 can be S(0) 2 NHMe.
  • R 2 is as defined in any one of claims 30-41.
  • R 2 is selected from the group consisting of: H, halo, cyano, OC(0)R 4B , and SR 5B (e.g., R 2 can be selected from H, halo, and cyano).
  • R 2 is halo (e.g., chloro).
  • R 2 is halo; and R 1 is S02R 4A or S(0)2N(R 6A )2.
  • R 2 can be chloro; and R 1 can be S02R 4A or S(0)2N(R 6A )2, wherein R 4A is Ci- 6 alkyl; and each R 6A is independently H or Ci-10 alkyl
  • R 1 and R 2 are as defined in any one of claims 42-45.
  • each of R 3 , R 4 , and R 5 is as defined in any one of claims 46-47.
  • R 3 is selected from the group consisting of:
  • halo e.g., F
  • Ci- 6 alkyl e.g., methyl
  • R 3 is H.
  • R 4 is selected from the group consisting of:
  • Ci- 6 alkoxy e.g., methoxy
  • R 4 is H.
  • R 5 is H.
  • each of R 3 and R 4 is H.
  • R 3 and R 4 is as defined in any one of claims 51-54.
  • the compound is selected from the group of compounds listed in Table 1 :
  • R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2; OR R 1 and R 2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(
  • a chemical entity e.g., a compound that inhibits (e.g., antagonizes) STING, or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination thereof
  • a pharmaceutical composition that includes the chemical entity and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein.
  • the chemical entities can be administered in combination with one or more conventional pharmaceutical excipients.
  • compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-
  • Cyclodextrins such as a-, b, and g-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl-P-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein.
  • Dosage forms or compositions containing a chemical entity as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared.
  • the contemplated compositions may contain 0.001%-100% of a chemical entity provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy , 22 nd Edition (Pharmaceutical Press, London, UK. 2012).
  • the chemical entities described herein or a pharmaceutical composition thereof can be administered to subject in need thereof by any accepted route of administration.
  • Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intracisternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, nasal, nasogastric
  • compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes.
  • parenteral administration e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes.
  • such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • injectables either as liquid solutions or suspensions
  • solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • the preparation of such formulations will be known to those of skill in the art in light of the present disclosure.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the 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 dispersion, and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p- oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylo
  • suppositories can be prepared by mixing the chemical entities described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound.
  • compositions for rectal administration are in the form of an enema.
  • the compounds described herein or a pharmaceutical composition thereof are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the chemical entity is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol mono
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a chemical entity provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like.
  • a diluent such as lactose, sucrose, dicalcium phosphate, or the like
  • a lubricant such as magnesium stearate or the like
  • a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like.
  • a powder, marume, solution or suspension (e.g, in propylene carbonate, vegetable oils, PEG’s, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule).
  • a capsule gelatin or cellulose base capsule.
  • Unit dosage forms in which one or more chemical entities provided herein or additional active agents are physically separated are also contemplated; e.g. , capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two- compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.
  • physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms.
  • Various preservatives are well known and include, for example, phenol and ascorbic acid.
  • the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient.
  • solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the chemical entity to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel.
  • Exemplary formulation techniques are described in, e.g., Filipski, K.J., et ak, Current Topics in Medicinal Chemistry, 2013, 13, 776-802, which is incorporated herein by reference in its entirety.
  • Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls.
  • Upper-GI targeting techniques e.g., Accordion Pill (Intec Pharma)
  • floating capsules e.g., floating capsules, and materials capable of adhering to mucosal walls.
  • enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid-methyl methacrylate copolymers), and Marcoat).
  • Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap.
  • Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, ETDA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxychloro complex; Allergan, Inc.)).
  • viscogens e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol
  • Stabilizers e.g., Pluronic (triblock copolymers), Cyclodextrins
  • Preservatives e.g., Benzalkonium chloride, ETDA, SofZ
  • Topical compositions can include ointments and creams.
  • Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives.
  • Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil.
  • Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase.
  • the oil phase also sometimes called the“internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant.
  • the emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant.
  • an ointment base should be inert, stable, nonirritating and non sensitizing.
  • compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers.
  • lipids interbilayer crosslinked multilamellar vesicles
  • biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles and nanoporous particle-supported lipid bilayers.
  • the dosages may be varied depending on the requirement of the patient, the severity of the condition being treating and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts.
  • the total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery.
  • the compounds described herein are administered at a dosage of from about 0.001 mg/Kg to about 500 mg/Kg (e.g., from about 0.001 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 150 mg/Kg; from about 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 50 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 5 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from about 0.01 mg/Kg to about 0.5 mg/Kg; from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0.
  • the foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month).
  • a daily basis e.g., as a single dose or as two or more divided doses
  • non-daily basis e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month.
  • the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more.
  • a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 4 months,
  • a therapeutic compound is administered to an individual for a period of time followed by a separate period of time.
  • a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped.
  • the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time.
  • a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days,
  • a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
  • methods for treating a subject having condition, disease or disorder in which increased (e.g., excessive) STING pathway activity, e.g., increased (e.g., excessive) STING or cGAS activity e.g., , e.g., STING or cGAS signaling
  • increased STING pathway activity e.g., increased (e.g., excessive) STING or cGAS activity
  • STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., immune disorders, cancer)
  • the condition, disease or disorder is cancer.
  • cancer include melanoma, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include breast cancer, colon cancer, rectal cancer, colorectal cancer, kidney or renal cancer, clear cell cancer lung cancer including small-cell lung cancer, non- small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, squamous cell cancer (e.g.
  • epithelial squamous cell cancer renal clear cell carcinoma, uveal melanoma, tongue squamous cell carcinoma, breast cancer, skin cancer, cervical cancer, ovarian cancer, prostate cancer, prostatic neoplasms, liver cancer, bladder cancer, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, gastrointestinal stromal tumor, pancreatic cancer, head and neck cancer, glioblastoma, retinoblastoma, astrocytoma, thecomas, arrhenoblastomas, hepatoma, hematologic malignancies including non-Hodgkins lymphoma (NHL), multiple myeloma, myelodysplasia disorders, myeloproliferative disorders, chronic myelogenous leukemia, and acute hematologic malignancies, endometrial or uterine carcinoma, endometriosis, endometrial stromal sarcoma, fibros
  • the condition, disease or disorder is a neurological disorder, which includes disorders that involve the central nervous system (brain, brainstem and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (parts of which are located in both central and peripheral nervous system).
  • a neurological disorder which includes disorders that involve the central nervous system (brain, brainstem and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (parts of which are located in both central and peripheral nervous system).
  • Non-limiting examples of cancer include acquired epileptiform aphasia; acute disseminated encephalomyelitis; adrenoleukodystrophy; age-related macular degeneration; agenesis of the corpus callosum; agnosia; Aicardi syndrome; Alexander disease; Alpers' disease; alternating hemiplegia; Alzheimer's disease; Vascular dementia; amyotrophic lateral sclerosis; anencephaly; Angelman syndrome; angiomatosis; anoxia; aphasia; apraxia; arachnoid cysts; arachnoiditis; Anronl-Chiari malformation; arteriovenous malformation; Asperger syndrome; ataxia tel egi ectasia; attention deficit hyperactivity disorder; autism; autonomic dysfunction; back pain; Batten disease; Behcet's disease; Bell's palsy; benign essential blepharospasm; benign focal; amyotrophy; benign intracranial hyper
  • the condition, disease or disorder is STING-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.
  • STING-associated conditions e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.
  • SAVI STING-associated vasculopathywith onset in infancy
  • AVS Aicardi-Goutieres Syndrome
  • genetic forms of lupus e.g., systemic
  • Non-limiting examples include rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel diseases (IBDs) comprising Crohn disease (CD) and ulcerative colitis (UC), which are chronic inflammatory conditions with polygenic susceptibility.
  • the condition is an inflammatory bowel disease.
  • the condition is Crohn’s disease, autoimmune colitis, iatrogenic autoimmune colitis, ulcerative colitis, colitis induced by one or more chemotherapeutic agents, colitis induced by treatment with adoptive cell therapy, colitis associated by one or more alloimmune diseases (such as graft-vs-host disease, e.g., acute graft vs.
  • the condition is alloimmune disease (such as graft-vs-host disease, e.g., acute graft vs. host disease and chronic graft vs.
  • celiac disease irritable bowel syndrome
  • rheumatoid arthritis lupus
  • scleroderma e.g., cutaneous T-cell lymphoma
  • uveitis e.g., uveitis
  • mucositis e.g., oral mucositis, esophageal mucositis or intestinal mucositis.
  • modulation of the immune system by STING provides for the treatment of diseases, including diseases caused by foreign agents.
  • exemplary infections by foreign agents which may be treated and/or prevented by the method of the present invention include an infection by a bacterium (e.g., a Gram-positive or Gram negative bacterium), an infection by a fungus, an infection by a parasite, and an infection by a virus.
  • the infection is a bacterial infection (e.g., infection by E.
  • the infection is a fungal infection (e.g. infection by a mould, a yeast, or a higher fungus).
  • the infection is a parasitic infection (e.g., infection by a single-celled or multicellular parasite, including Giardia duodenalis, Cryptosporidium parvum, Cyclospora cayetanensis, and Toxoplasma gondiz).
  • the infection is a viral infection (e.g., infection by a virus associated with AIDS, avian flu, chickenpox, cold sores, common cold, gastroenteritis, glandular fever, influenza, measles, mumps, pharyngitis, pneumonia, rubella, SARS, and lower or upper respiratory tract infection (e.g., respiratory syncytial virus)).
  • a viral infection e.g., infection by a virus associated with AIDS, avian flu, chickenpox, cold sores, common cold, gastroenteritis, glandular fever, influenza, measles, mumps, pharyngitis, pneumonia, rubella, SARS, and lower or upper respiratory tract infection (e.g., respiratory syncytial virus)).
  • condition, disease or disorder is hepatits B (see, e.g., WO 2015/061294).
  • the condition, disease or disorder is selected from cardiovascular diseases (including e.g., myocardial infarction).
  • the condition, disease or disorder is age-related macular degeneration.
  • condition, disease or disorder is mucositis, also known as stomatitits, which can occur as a result of chemotherapy or radiation therapy, either alone or in combination as well as damage caused by exposure to radiation outside of the context of radiation therapy.
  • the condition, disease or disorder is uveitis, which is inflammation of the uvea (e.g., anterior uveitis, e.g., iridocyclitis or LTDis; intermediate uveitis (also known as pars planitis); posterior uveitis; or chorioretinitis, e.g., pan-uveitis).
  • uveitis inflammation of the uvea
  • anterior uveitis e.g., iridocyclitis or ulceris
  • intermediate uveitis also known as pars planitis
  • posterior uveitis e.g., pan-uveitis
  • chorioretinitis e.g., pan-uveitis
  • the condition, disease or disorder is selected from the group consisting of a cancer, a neurological disorder, an autoimmune disease, hepatitis B, uvetitis, a cardiovascular disease, age-related macular degeneration, and mucositis.
  • Still other examples can include those indications discussed herein and below in contemplated combination therapy regimens.
  • This disclosure contemplates both monotherapy regimens as well as combination therapy regimens.
  • the methods described herein can further include administering one or more additional therapies (e.g., one or more additional therapeutic agents and/or one or more therapeutic regimens) in combination with administration of the compounds described herein.
  • the methods described herein can further include administering one or more additional cancer therapies.
  • the one or more additional cancer therapies can include, without limitation, surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy, cancer vaccines (e.g., HPV vaccine, hepatitis B vaccine, Oncophage, Provenge) and gene therapy, as well as combinations thereof.
  • Immunotherapy including, without limitation, adoptive cell therapy, the derivation of stem cells and/or dendritic cells, blood transfusions, lavages, and/or other treatments, including, without limitation, freezing a tumor.
  • the one or more additional cancer therapies is chemotherapy, which can include administering one or more additional chemotherapeutic agents.
  • the additional chemotherapeutic agent is an immunomodulatory moiety, e.g., an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD-1 - PD-
  • L2 interleukin-2 (IL-2), indoleamine 2,3 -di oxygenase (IDO), IL-10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR,
  • the immune checkpoint inhibitor is selected from the group consisting of: Urelumab, PF-05082566, MEDI6469, TRX518, Varlilumab, CP-870893, Pembrolizumab (PD1), Nivolumab (PD1), Atezolizumab (formerly
  • MPDL3280A (PDL1), MEDI4736 (PD-L1), Avelumab (PD-L1), PDR001 (PD1), BMS-986016, MGA271, Lirilumab, IPH2201, Emactuzumab, INCB024360, Galunisertib,
  • the additional chemotherapeutic agent is an alkylating agent.
  • Alkylating agents are so named because of their ability to alkylate many nucleophilic functional groups under conditions present in cells, including, but not limited to cancer cells.
  • an alkylating agent includes, but is not limited to, Cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin.
  • alkylating agents can function by impairing cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules or they can work by modifying a cell's DNA.
  • an alkylating agent is a synthetic, semisynthetic or derivative.
  • the additional chemotherapeutic agent is an anti metabolite.
  • Anti-metabolites masquerade as purines or pyrimidines, the building-blocks of DNA and in general, prevent these substances from becoming incorporated in to DNA during the "S" phase (of the cell cycle), stopping normal development and division.
  • Anti- metabolites can also affect RNA synthesis.
  • an antimetabolite includes, but is not limited to azathioprine and/or mercaptopurine.
  • an anti metabolite is a synthetic, semisynthetic or derivative.
  • the additional chemotherapeutic agent is a plant alkaloid and/or terpenoid.
  • alkaloids are derived from plants and block cell division by, in general, preventing microtubule function.
  • a plant alkaloid and/or terpenoid is a vinca alkaloid, a podophyllotoxin and/or a taxane.
  • Vinca alkaloids in general, bind to specific sites on tubulin, inhibiting the assembly of tubulin into microtubules, generally during the M phase of the cell cycle.
  • a vinca alkaloid is derived, without limitation, from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea).
  • a vinca alkaloid includes, without limitation, Vincristine, Vinblastine, Vinorelbine and/or Vindesine.
  • a taxane includes, but is not limited, to Taxol, Paclitaxel and/or Docetaxel.
  • a plant alkaloid or terpernoid is a synthetic, semisynthetic or derivative.
  • a podophyllotoxin is, without limitation, an etoposide and/or teniposide.
  • a taxane is, without limitation, docetaxel and/or ortataxel. [021]
  • a cancer therapeutic is a topoisom erase.
  • Topoisom erases are essential enzymes that maintain the topology of DNA. Inhibition of type I or type II topoisomerases interferes with both transcription and replication of DNA by upsetting proper DNA supercoiling.
  • a topoisomerase is, without limitation, a type I topoisomerase inhibitor or a type II topoisomerase inhibitor.
  • a type I topoisomerase inhibitor is, without limitation, a camptothecin.
  • a camptothecin is, without limitation, exatecan, irinotecan, lurtotecan, topotecan, BNP 1350, CKD 602, DB 67 (AR67) and/or ST 1481.
  • a type II topoisomerase inhibitor is, without limitation, epipodophyllotoxin.
  • an epipodophyllotoxin is, without limitation, an amsacrine, etoposid, etoposide phosphate and/or teniposide.
  • a topoisomerase is a synthetic, semisynthetic or derivative, including those found in nature such as, without limitation, epipodophyllotoxins, substances naturally occurring in the root of American Mayapple (Podophyllum peltatum).
  • the additional chemotherapeutic agent is a stilbenoid.
  • a stilbenoid includes, but is not limited to, Resveratrol, Piceatannol, Pinosylvin, Pterostilbene, Alpha- Viniferin, Ampelopsin A, Ampelopsin E, Diptoindonesin C, Diptoindonesin F, Epsilon- Vinferin, Flexuosol A, Gnetin H, Hemsleyanol D, Hopeaphenol, Trans-Diptoindonesin B, Astringin, Piceid and Diptoindonesin A.
  • a stilbenoid is a synthetic, semisynthetic or derivative.
  • the additional chemotherapeutic agent is a cytotoxic antibiotic.
  • a cytotoxic antibiotic is, without limitation, an actinomycin, an anthracenedione, an anthracycline, thalidomide, dichloroacetic acid, nicotinic acid, 2- deoxyglucose and/or chlofazimine.
  • an actinomycin is, without limitation, actinomycin D, bacitracin, colistin (polymyxin E) and/or polymyxin B.
  • an antracenedione is, without limitation, mitoxantrone and/or pixantrone.
  • an anthracycline is, without limitation, bleomycin, doxorubicin (Adriamycin), daunorubicin (daunomycin), epirubicin, idarubicin, mitomycin, plicamycin and/or valrubicin.
  • a cytotoxic antibiotic is a synthetic, semi sy ntheti c or derivative .
  • the additional chemotherapeutic agent is selected from endostatin, angiogenin, angiostatin, chemokines, angioarrestin, angiostatin (plasminogen fragment), basement-membrane collagen-derived anti-angiogenic factors (tumstatin, canstatin, or arrestin), anti-angiogenic antithrombin III, signal transduction inhibitors, cartilage-derived inhibitor (CDI), CD59 complement fragment, fibronectin fragment, gro- beta, heparinases, heparin hexasaccharide fragment, human chorionic gonadotropin (hCG), interferon alpha/beta/gamma, interferon inducible protein (IP- 10), interleukin- 12, kringle 5 (plasminogen fragment), metalloproteinase inhibitors (TIMPs), 2-methoxyestradiol, placental ribonuclease inhibitor, plasminogen activator inhibitor, platelet factor-4 (PF4), prolactin, cartilage
  • the additional chemotherapeutic agent is selected from abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly-l-Lproline-t- butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3',4'-didehydro-4'- deoxy-8'-norvin-caleukoblastine, docetaxol, doxetaxel, cyclophosphamide, carboplatin, carmustine, cisplatin, cryptophycin,
  • the additional chemotherapeutic agent is platinum, cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, azathioprine, mercaptopurine, vincristine, vinblastine, vinorelbine, vindesine, etoposide and teniposide, paclitaxel, docetaxel, irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide, 5-fluorouracil, leucovorin, methotrexate, gemcitabine, taxane, leucovorin, mitomycin C, tegafur-uracil, idarubicin, fludarabine, mitoxantrone, ifosfamide and doxorubicin.
  • Additional agents include inhibitors of mTOR (mammalian target of rapamycin), including but
  • the additional chemotherapeutic agent can be selected from those delineated in U.S. Patent 7,927,613, which is incorporated herein by reference in its entirety.
  • the additional therapeutic agent and/or regimen are those that can be used for treating other STING-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis and the like.
  • STING-associated conditions e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheuma
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating rheumatoid arthritis include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), corticosteroids (e.g, prednisone), disease-modifying antirheumatic drugs (DMARDs; e.g., methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), leflunomide (Arava®), hydroxychloroquine (Plaquenil), PF-06650833, iguratimod, tofacitinib (Xeljanz®), ABBV-599, evobrutinib, and sulfasalazine (Azulfidine®)), and biologies (e.g., abatacept (Orencia®), adalimumab (Humira®), anakinra (Kineret
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating lupus include steroids, topical immunomodulators (e.g., tacrolimus ointment (Protopic®) and pimecrolimus cream (Elidel®)), thalidomide (Thalomid®), non-steroidal anti inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), antimalarial drugs (e.g., Hydroxychloroquine (Plaquenil)), corticosteroids (e.g, prednisone) and immunomodulators (e.g., evobrutinib, iberdomide, voclosporin, cenerimod, azathioprine (Imuran®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral, Sandimmune®, Gengraf®), and mycophenolate mofetil) baricitinb
  • non-limiting treatments for systemic lupus erythematosus include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), antimalarial drugs (e.g., Hydroxychloroquine (Plaquenil)), corticosteroids (e.g, prednisone) and immunomodulators (e.g., iberdomide, voclosporin, azathioprine (Imuran®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral, Sandimmune®, Gengraf®), and mycophenolate mofetil, baricitinb, filogotinib, and PF-06650833), and biologies (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, vobarilizumab
  • non-limiting examples of treatments for cutaneous lupus include steroids, immunomodulators (e.g., tacrolimus ointment (Protopic®) and pimecrolimus cream (Elidel®)), GS-9876, filogotinib, and thalidomide (Thalomid®).
  • agents and regimens for treating drug-induced and/or neonatal lupus can also be administered.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating STING-associated vasculopathy with onset in infancy include JAK inhibitors (e.g., tofacitinib, ruxolitinib, filgotinib, and baricitinib).
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating Aicardi-Goutieres Syndrome include physiotherapy, treatment for respiratory complications, anticonvulsant therapies for seizures, tube-feeding, nucleoside reverse transcriptase inhibitors (e.g., emtricitabine (e.g., Emtriva®), tenofovir (e.g., Viread®), emtricitabine/tenofovir (e.g., Truvada®), zidovudine, lamivudine, and abacavir), and JAK inhibitors (e.g., tofacitinib, ruxolitinib, filgotinib, and baricitinib).
  • nucleoside reverse transcriptase inhibitors e.g., emtricitabine (e.g., Emtriva®), tenofovir (e.g., Viread®), emtricitabine/tenofovir (e.g., Tru
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating IBDs include 6-mercaptopurine, AbGn-168H, ABX464, ABT-494, adalimumab, AJM300, alicaforsen, AMG139, anrukinzumab, apremilast, ATR-107 (PF0530900), autologous CD34-selected peripheral blood stem cells transplant, azathioprine, bertilimumab, BI 655066, BMS-936557, certolizumab pegol (Cimzia®), cobitolimod, corticosteroids (e.g., prednisone, Methylprednisolone, prednisone), CP-690,550, CT-P13, cyclosporine, DIMS0150, E6007, E6011, etrasimod, etrolizumab, fecal microbial transplantation, figlotinib, fmgolimod,
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating irritable bowel syndrome include alosetron, bile acid sequesterants (e.g., cholestyramine, colestipol, colesevelam), chloride channel activators (e.g., lubiprostone), coated peppermint oil capsules, desipramine, dicyclomine, ebastine, eluxadoline, famesoid X receptor agonist (e.g., obeticholic acid), fecal microbiota transplantation, fluoxetine, gabapentin, guanylate cyclase-C agonists (e.g., linaclotide, plecanatide), ibodutant, imipramine, JCM- 16021, loperamide, lubiprostone, nortriptyline, ondansetron, opioids, paroxetine, pinaverium, polyethylene glycol, pregabalin, probiotics, ramosetron,
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating scleroderma include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), corticosteroids (e.g, prednisone), immunomodulators (e.g., azathioprine, methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral®, Sandimmune®, Gengraf®), antithymocyte globulin, mycophenolate mofetil, intravenous immunoglobulin, rituximab, sirolimus, and alefacept), calcium channel blockers (e.g., nifedipine), alpha blockers, serotonin receptor antagonists, angiotensin II receptor inhibitors, statins, local
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating Crohn’s Disease include adalimumab, autologous CD34-selected peripheral blood stem cells transplant, 6-mercaptopurine, azathioprine, certolizumab pegol (Cimzia®), corticosteroids (e.g., prednisone), etrolizumab, E6011, fecal microbial transplantation, figlotinib, guselkumab, infliximab, IL-2, JAK inhibitors, matrix metalloproteinase 9 (MMP 9) inhibitors (e.g., GS-5745), MEDI2070, mesalamine, methotrexate, natalizumab, ozanimod, RHB-104, rifaximin, risankizumab, SHP647, sulfasalazine, thalidomide, upadacitinib, V
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating UC include AbGn-168H, ABT-494, ABX464, apremilast, PF-00547659, PF-06687234, 6- mercaptopurine, adalimumab, azathioprine, bertilimumab, brazikumab (MEDI2070), cobitolimod, certolizumab pegol (Cimzia®), CP-690,550, corticosteroids (e.g., multimax budesonide, Methylprednisolone), cyclosporine, E6007, etrasimod, etrolizumab, fecal microbial transplantation, figlotinib, guselkumab, golimumab, IL-2, IMU-838, infliximab, matrix metalloproteinase 9 (MMP9) inhibitors (e.g., GS-57
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating autoimmune colitis include corticosteroids (e.g., budesonide, prednisone, prednisolone, Beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, mesalamine, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating iatrogenic autoimmune colitis include corticosteroids (e.g., budesonide, prednisone, prednisolone, Beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis induced by one or more chemotherapeutics agents include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, mesalamine, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis induced by treatment with adoptive cell therapy include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
  • corticosteroids e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate
  • diphenoxylate/atropine e.g., infliximab
  • loperamide e.g., loperamide
  • TIP60 inhibitors see, e.g., U.S. Patent Application Publication No. 2012/0202848
  • vedolizumab e.g.,
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis associated with one or more alloimmune diseases include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), sulfasalazine, and eicopentaenoic acid.
  • corticosteroids e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate
  • sulfasalazine eicopentaenoic acid.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating radaiation enteritis include teduglutide, amifostine, angiotensin-converting enzyme (ACE) inhibitors (e.g., benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, and trandolapril), probiotics, selenium supplementation, statins (e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, and pitavastatin), sucralfate, and vitamin E.
  • ACE angiotensin-converting enzyme
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating collagenous colitis include 6-mercaptopurine, azathaioprine, bismuth subsalicate, Boswellia serrata extract, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), loperamide, mesalamine, methotrexate, probiotics, and sulfasalazine.
  • corticosteroids e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate
  • loperamide mesalamine, methotrexate, probiotics, and sulfasalazine.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating lyphocytic colitis include 6-mercaptopurine, azathioprine, bismuth subsalicylate, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), loperamide, mesalamine, methotrexate, and sulfasalazine.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating microscopic colitis include 6-mercaptopurine, azathioprine, bismuth subsalicylate, Boswellia serrata extract, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), fecal microbial transplantation, loperamide, mesalamine, methotrexate, probiotics, and sulfasalazine.
  • corticosteroids e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate
  • corticosteroids e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate
  • fecal microbial transplantation loperamide, mesalamine, methot
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating alloimmune disease include intrauterine platelet transfusions, intravenous immunoglobin, maternal steroids, abatacept, alemtuzumab, alphal -antitrypsin, AMG592, antithymocyte globulin, barcitinib, basiliximab, bortezomib, brentuximab, cannabidiol, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, glasdegib, ibrutinib, IL-2, infliximab, itacitinib, LBH589, maraviroc, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, pevonedistat, photobiomodulation,
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating multiple sclerosis include alemtuzumab (Lemtrada®), ALKS 8700, amiloride, ATX- MS-1467, azathioprine, baclofen (Lioresal®), beta interferons (e.g., IFN-b- I a, IFN-b- 1 b), cladribine, corticosteroids (e.g., methylprednisolone), daclizumab, dimethyl fumarate (Tecfidera®), fmgolimod (Gilenya®), fluoxetine, glatiramer acetate (Copaxone®), hydroxychloroquine, ibudilast, idebenone, laquinimod, lipoic acid, losartan, masitinib, MD1003 (biotin), mitoxantrone, montelukast, natalizumab (Tysabri®),
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating graft-vs-host disease include abatacept, alemtuzumab, alphal -antitrypsin, AMG592, antithymocyte globulin, barcitinib, basiliximab, bortezomib, brentuximab, cannabidiol, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, glasdegib, ibrutinib, IL-2, imatinib, infliximab, itacitinib, LBH589, maraviroc, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, pevonedistat, photobiomodulation, photopheresis, rux
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating acute graft-vs-host disease include alemtuzumab, alpha- 1 antitrypsin, antithymocyte globulin, basiliximab, brentuximab, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, ibrutinib, infliximab, itacitinib, LBH589, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, photopheresis, ruxolitinib, sirolimus, tacrolimus, and tocilizumab.
  • corticosteroids e.g., methylprednisone, prednisone
  • cyclosporine e.g.,
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating chronic graft vs. host disease include abatacept, alemtuzumab, AMG592, antithymocyte globulin, basiliximab, bortezomib, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, denileukin diftitox, glasdegib, ibrutinib, IL-2, imatinib, infliximab, mycophenolate mofetil, pentostatin, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.
  • corticosteroids e.g., methylprednisone, prednisone
  • corticosteroids e.g., methylpred
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating celiac disease include AMG 714, AMY01, Aspergillus niger prolyl endoprotease, BL- 7010, CALY-002, GBR 830, Hu-Mik-Beta-1, IMGX003, KumaMax, Larazotide Acetate, Nexvan2®, pancrelipase, TIMP-GLIA, vedolizumab, and ZED1227.
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating psoriasis include topical corticosteroids, topical crisaborole/AN2728, topical SNA-120, topical SAN021, topical tapinarof, topical tocafmib, topical IDP-118, topical M518101, topical calcipotriene and betamethasone dipropionate (e.g., MC2-01 cream and Taclonex®), topical P-3073, topical LEO 90100 (Enstilar®), topical betamethasone dipropriate (Semivo®), halobetasol propionate (Ultravate®), vitamin D analogues (e.g., calcipotriene (Dovonex®) and calcitriol (Vectical®)), anthralin (e.g., Dritho-scalp® and Dritho-creme®), topical retinoids (e.g., t
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating cutaneous T-cell lymphoma include phototherapy (e.g., exposure to sunlight, UVB phototherapy, narrow band UVB phototherapy, Goeckerman therapy, psoralen plus ultraviolet A (PUVA) therapy, and excimer laser), extracorporeal photopheresis, radiation therapy (e.g., spot radiation and total skin body electron beam therapy), stem cell transplant, corticosteroids, imiquimod, bexarotene gel, topical bis-chloroethyl-nitrourea, mechlorethamine gel, vorinostat (Zolinza®), romidepsin (Istodax®), pralatrexate (Folotyn®) biologies (e.g., alemtuzumab (Campath®), brentuximab vedotin (SGN-35), mogamulizumab, and IPH4102).
  • phototherapy e.g., exposure
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating uveitis include corticosteroids (e.g., intravitreal triamcinolone acetonide injectable suspensions), antibiotics, antivirals (e.g., acyclovir), dexamethasone, immunomodulators (e.g., tacrolimus, leflunomide, cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral®, Sandimmune®, Gengraf®), chlorambucil, azathioprine, methotrexate, and mycophenolate mofetil), biologies (e.g., infliximab (Remicade®), adalimumab (Humira®), etanercept (Enbrel®), golimumab (Simponi®), certolizumab (Cimzia®), rituximab (Ritux
  • Non-limiting examples of additional therapeutic agents and/or regimens for treating mucositis include AGO 13, SGX942 (dusquetide), amifostine (Ethyol®), cryotherapy, cepacol lonzenges, capsaicin lozenges, mucoadhesives (e.g., MuGard®) oral diphenhydramine (e.g., Benadry® elixir), oral bioadherents (e.g., polyvinylpyrrolidone- sodium hyaluronate gel (Gelclair®)), oral lubricants (e.g., Oral Balance®), caphosol, chamomilla recutita mouthwash, edible grape plant exosome, antiseptic mouthwash (e.g., chlorhexidine gluconate (e.g., Peridex® or Periogard®), topical pain relievers (e.g., lidocaine, benzocaine, dyclonine hydroch
  • non limiting examples of treatments for oral mucositis include AG013, amifostine (Ethyol®), cryotherapy, cepacol lonzenges, mucoadhesives (e.g., MuGard®) oral diphenhydramine (e.g., Benadry® elixir), oral bioadherents (e.g., polyvinylpyrrolidone-sodium hyaluronate gel (Gelclair®)), oral lubricants (e.g., Oral Balance®), caphosol, chamomilla recutita mouthwash, edible grape plant exosome, antiseptic mouthwash (e.g., chlorhexidine gluconate (e.g., Peridex® or Periogard®), topical pain relievers (e.g., lidocaine, benzocaine, dyclonine hydrochloride, xylocaine (e.g., viscous xylocaine (e
  • non-limiting examples of treatments for esophageal mucositis include xylocaine (e.g., gel viscous Xylocaine 2%).
  • treatments for intestinal mucositis, treatments to modify intestinal mucositis, and treatments for intestinal mucositis signs and symptoms include gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)).
  • an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox)
  • an antifungal e.g., nystatin
  • an analgesic e.g., hurricane liquid
  • the second therapeutic agent or regimen is administered to the subject prior to contacting with or administering the chemical entity (e.g., about one hour prior, or about 6 hours prior, or about 12 hours prior, or about 24 hours prior, or about 48 hours prior, or about 1 week prior, or about 1 month prior).
  • the chemical entity e.g., about one hour prior, or about 6 hours prior, or about 12 hours prior, or about 24 hours prior, or about 48 hours prior, or about 1 week prior, or about 1 month prior.
  • the second therapeutic agent or regimen is administered to the subject at about the same time as contacting with or administering the chemical entity.
  • the second therapeutic agent or regimen and the chemical entity are provided to the subject simultaneously in the same dosage form.
  • the second therapeutic agent or regimen and the chemical entity are provided to the subject concurrently in separate dosage forms.
  • the second therapeutic agent or regimen is administered to the subject after contacting with or administering the chemical entity (e.g., about one hour after, or about 6 hours after, or about 12 hours after, or about 24 hours after, or about 48 hours after, or about 1 week after, or about 1 month after).
  • the chemical entity e.g., about one hour after, or about 6 hours after, or about 12 hours after, or about 24 hours after, or about 48 hours after, or about 1 week after, or about 1 month after.
  • the methods described herein further include the step of identifying a subject (e.g., a patient) in need of such treatment (e.g., by way of biopsy, endoscopy, or other conventional method known in the art).
  • the STING protein can serve as a biomarker for certain types of cancer, e.g., colon cancer and prostate cancer.
  • identifying a subject can include assaying the patient’s tumor microenvironment for the absence of T-cells and/or presence of exhausted T-cells, e.g., patients having one or more cold tumors. Such patients can include those that are resistant to treatment with checkpoint inhibitors.
  • such patients can be treated with a chemical entity herein, e.g., to recruit T-cells into the tumor, and in some cases, further treated with one or more checkpoint inhibitors, e.g., once the T-cells become exhausted.
  • a chemical entity herein e.g., to recruit T-cells into the tumor
  • one or more checkpoint inhibitors e.g., once the T-cells become exhausted.
  • the chemical entities, methods, and compositions described herein can be administered to certain treatment-resistant patient populations (e.g., patients resistant to checkpoint inhibitors; e.g., patients having one or more cold tumors, e.g., tumors lacking T-cells or exhausted T-cells).
  • certain treatment-resistant patient populations e.g., patients resistant to checkpoint inhibitors; e.g., patients having one or more cold tumors, e.g., tumors lacking T-cells or exhausted T-cells.
  • Amine (e.g., 1-2) (1.0 eq.) and carboxylic acid (e.g., 1-1) (1.0 eq.) are dissolved in 2 ml DMF.
  • 5.0 eq. of triethylamine and 1.0 Eq. of EDC is added.
  • the resulting mixture is stirred for 24h, after which it is poured onto a mixture of DCM and 10% citric acid (1 : 1).
  • the phases are separated, and the aqueous phase is extracted with DCM.
  • the combined organic phases are washed with 10 mL water, dried over MgSCE, and concentrated under vacuum.
  • the solid is dissolved in DCM and adsorbed on 1.2g Silica, followed by flash chromatography (12 g S1O2, elution with hexane/ AcOEt) to yield the purified compound.
  • amine e.g., 1-1
  • acid chloride e.g. 1-3
  • EDC e.g. 1,3-bis(triethyl)
  • the mixture stirred for 24h.
  • the mixture is poured onto a mixture of DCM and 10% citric acid (1 : 1).
  • the phases are separated, and the aqueous phase is extracted with DCM.
  • the combined organic phases are washed with 10 ml water, dried over MgSCri, and concentrated under vacuum.
  • DIEA N,N-diisopropylethylamine
  • HATU 2-(7-azabenzotriazol-l-yl)-N,N,N , ,N’-tetramethyluronium hexafluorophosphate
  • TBS tertbutylaimethylsilyl chloride
  • NEE ammonia
  • n-Bu n-butyl
  • Ph phenyl
  • the progress of reactions was often monitored by TLC or LC-MS.
  • the identity of the products was often confirmed by LC-MS.
  • the LC-MS was recorded using one of the following methods.
  • Method A Titank C18, 50x3 mm, 3 um column, 0.3 uL injection, 1.5 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection.
  • Mobile phase A Water+5mMNH4HC03 and Mobile Phase B: Acetonitrile. 10% MPB to 95.0% in 1.39 min, hold at 95% MPB for 0.8 min, 95% MPB to 10% in 0.03 min, then equilibration to 10% MPB for 0.27 min.
  • Method B EVO-C18, 50x3 mm, 2.6 um column, 2.0 uL injection, 1.2 mL/min flow rate, 90-900 amu scan range, 254 nm UV detection.
  • Mobile phase A Water+5mMNH4HC03 and Mobile Phase B: Acetonitrile. 10% MPB to 95.0% in 1.99 min, hold at 95% MPB for 0.6 min, 95% MPB to 10% in 0.20 min, then equilibration to 10% MPB for 0.25 min.
  • Method D Poroshell HPH-C18, 50 *3mm, 2.7 pL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection.
  • Mobile Phase A (MPA) Water/0.04% NLLOH and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 1.99 min, hold at 95% MPB for 0.6 min, 95% MPB to 10% in 0.03 min, then equilibration to 10% MPB for 0.17 min.
  • Method L Shim-pack XR-ODS, 50 *3.0 mm, 50 *3.0 mm, 2.2 um column, 2.2 pL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection.
  • Mobile Phase A (MPA): Water/0.05% TFA and Mobile Phase B (MPB): Acetonitrile. Elution 5% MPB to 95% in 2 min, hold at 95% MPB for 0.7 min, 95% MPB to 10% in 0.05 min, then equilibration to 5% MPB for 0.25 min.
  • the final targets were purified by Prep-HPLC.
  • the Prep-HPLC was carried out using the following method.
  • Method I Pre-HPLC: Column: Xselect CSH OBD Column 30* 150mm 5um; Mobile Phase: Water (10MMOL/L NH HCC>3+0.1%NH3.H2O) and ACN, UV detection 254/210 nm.
  • Method K Pre-HPLC: Column: Atlantis HILIC OBD Column, 19* 150mm*5um; Mobile Phase: Water(10MMOL/L MLHCOs+O. U/oML.HiO) and ACN, UV detection 254/210 nm.
  • NMR was recorded on BRUKER NMR 300.03 Mz, DUL-C-H, ULTRASHIELDTM 300, AVANCE II 300 B-ACSTM 120 or BRUKER NMR 400.13 Mz, BBFO, ULTRASHIELDTM 400, AVANCE III 400, B-ACSTM 120.
  • Step 1 Synthesis of methyl 3-carbamoyl-4-chlorobenzoate
  • Step 2 Synthesis of tert-butyl (5-butylpyridin-2-yl)carbamate
  • THPl-DualTM KO-IFNAR2 Cells (obtained from invivogen) were maintained in RPMI, 10% FCS, 5 ml P/S, 2mM L-glut, lOmM Hepes, and 1 mM sodium pyruvate. Compounds were spotted in empty 384 well tissue culture plates (Greiner 781182) by Echo for a final concentration of 0.0017 - 100 mM Cells were plated into the TC plates at 40 pL per well, 2> ⁇ 10E6 cells/mL. For activation with STING ligand, 2'3'cGAMP (MW 718.38, obtained from Invivogen), was prepared in Optimem media.
  • Luciferase reporter assay 10 pL of supernatant from the assay was transferred to white 384-plate with flat bottom and squared wells. One pouch of QUANTI-LucTM Plus was dissolved in 25 mL of water. 100 pL of QLC Stabilizer per 25 mL of QUANTI- LucTM Plus solution was added. 50 pL of QUANTI-LucTM Plus/QLC solution per well was then added. Luminescence was measured on a Platereader (e.g., Spectramax I3X (Molecular Devices GF3637001)).
  • a Platereader e.g., Spectramax I3X (Molecular Devices GF3637001)
  • Luciferase reporter activity was then measured. ECso values were calculated by using standard methods known in the art.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes ("STING") or inhibit cyclic GMP-AMP Synthase ("cGAS"). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.

Description

Compounds and Compositions for Treating Conditions Associated with STING Activity
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Serial No. 62/769,500, filed on November 19, 2018; and U.S. Provisional Application Serial No.
62/861,108, filed on June 13, 2019; each of which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes (“STING”) or inhibit cyclic GMP-AMP Synthase (“cGAS”). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.
BACKGROUND
The cGAS/STING pathway is a component of inflammatory signaling pathways. When DNA is present in the cytosol of a cell, cGAS binds it and generates 2’-5’ cyclic GMP-AMP (cGAMP). Activated by cGAMP, STING induces the phosphorylation of and nuclear translocation of interferon (IFN) regulatory factors (IRFs). As transcription factors, IRFs regulate the expression of genes, including the type I IFNs, which regulate the activity of the immune system. STING, also known as transmembrane protein 173 (TMEM173) and
MPYS/MITA/ERIS, is a protein that in humans is encoded by the TMEM173 gene. STING has been shown to play a role in innate immunity. STING induces type I interferon production when cells are infected with intracellular pathogens, such as viruses, mycobacteria and intracellular parasites. Type I interferon, mediated by STING, protects infected cells and nearby cells from local infection in an autocrine and paracrine manner.
The STING pathway is pivotal in mediating the recognition of cytosolic DNA. In this context, STING, a transmembrane protein localized to the endoplasmic reticulum (ER), acts as a second messenger receptor for 2', 3' cyclic GMP-AMP (hereafter cGAMP), which is produced by cGAS after dsDNA binding. In addition, STING can also function as a primary pattern recognition receptor for bacterial cyclic dinucleotides (CDNs) and small molecule agonists. The recognition of endogenous or prokaryotic CDNs proceeds through the carboxy-terminal domain of STING, which faces into the cytosol and creates a V-shaped binding pocket formed by a STING homodimer. Ligand-induced activation of STING triggers its re-localization to the Golgi, a process essential to promote the interaction of STING with TBK1. This protein complex, in turn, signals through the transcription factors IRF-3 to induce type I interferons (IFNs) and other co-regulated antiviral factors. In addition, STING was shown to trigger NF-KB and MAP kinase activation. Following the initiation of signal transduction, STING is rapidly degraded, a step considered important in terminating the inflammatory response.
Excessive activation of STING is associated with a subset of monogenic autoinflammatory conditions, the so-called type I interferonopathies. Examples of these diseases include a clinical syndrome referred to as STING-associated vasculopathy with onset in infancy (SAVI), which is caused by gain-of-function mutations in TMEM173 (the gene name of STING). Moreover, STING is implicated in the pathogenesis of Aicardi- Goutieres Syndrome (AGS) and genetic forms of lupus. As opposed to SAVI, it is the dysregulation of nucleic acid metabolism that underlies continuous innate immune activation in AGS. Apart from these genetic disorders, emerging evidence points to a more general pathogenic role for STING in a range of inflammation-associated disorders such as systemic lupus erythematosus, rheumatoid arthritis and cancer. Thus, small molecule- based pharmacological interventions into the STING signaling pathway hold significant potential for the treatment of a wide spectrum of diseases
SUMMARY
This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes (“STING”) or inhibit cyclic GMP-AMP Synthase (“cGAS”). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.
As used herein, the term“STING antagonist” is an agent that decreases one or both of (i) the activity of STING (e.g., any of the exemplary activities of STING described herein) (e.g., as compared to the level of STING activity in the absence of the agent) and (ii) the expression level of STING in a mammalian cell (e.g., using any of the exemplary methods of detection described herein) (e.g., as compared to the expression level of STING in a mammalian cell not contacted with the agent) .An "antagonist" of STING also includes compounds that, at the protein level, directly bind or modify STING such that an activity of STING is decreased, e.g., by inhibition, blocking or dampening agonist-mediated responses, altered distribution, or otherwise. STING antagonists include chemical entities, which interfere or inhibit STING signaling.
As used herein, the term“cGAS inhibitor” is an agent that decreases one or both of (i) the activity of cGAS (e.g., any of the exemplary activities of cGAS described herein) (e.g., as compared to the level of cGAS activity in the absence of the agent) and (ii) the expression level of cGAS in a mammalian cell (e.g., using any of the exemplary methods of detection described herein) (e.g., as compared to the expression level of cGAS in a mammalian cell not contacted with the agent). An "inhibitor" of cGAS also includes compounds that, at the protein level, directly bind or modify cGAS such that an activity of cGAS is decreased, e.g., by inhibition, blocking or dampening agonist-mediated responses, altered distribution, or otherwise. cGAS inhibitors include chemical entities, which interfere or inhibit cGAS signaling.
Figure imgf000005_0001
In which R1, R2, R3, R4, R5, W, Q, and A can be as defined anywhere herein.
In one aspect, pharmaceutical compositions are featured that include a chemical entity described herein (e.g., a formula (I) compound described genetically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) and one or more pharmaceutically acceptable excipients.
In one aspect, methods for inhibiting the STING pathway, e.g., inhibiting (e.g., antagonizing) STING activity or inhibiting cGAS activity are featured that include contacting STING or cGAS with a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same). Methods include in vitro methods, e.g., contacting a sample that includes one or more cells comprising STING or cGAS (e.g., innate immune cells, e.g., mast cells, macrophages, dendritic cells (DCs), and natural killer cells) with the chemical entity. Methods can also include in vivo methods; e.g., administering the chemical entity to a subject (e.g., a human) having a disease in which increased (e.g., excessive) STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the disease.
In one aspect, methods of treating a condition, disease or disorder ameliorated by antagonizing STING or inhibiting cGAS are featured, e.g., treating a condition, disease or disorder in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). The methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
In another aspect, methods of treating cancer are featured that include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
In a further aspect, methods of treating other STING or cGAS-associated conditions are featured, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis. The methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
In another aspect, methods of suppressing STING-dependent type I interferon production in a subject in need thereof are featured that include administering to the subject an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same). In a further aspect, methods of treating a disease in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the disease are featured. The methods include administering to a subject in need of such treatment an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same).
In another aspect, methods of treatment are featured that include administering an effective amount of a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same) to a subject; wherein the subject has (or is predisposed to have) a disease in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the disease .
In a further aspect, methods of treatment that include administering to a subject a chemical entity described herein (e.g., a formula (I) compound described generically or specifically herein or a pharmaceutically acceptable salt thereof or compositions containing the same), wherein the chemical entity is administered in an amount effective to treat a disease in which increased (e.g., excessive) STING or cGAS activation (e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.
Embodiments can include one or more of the following features.
The chemical entity can be administered in combination with one or more additional therapeutic agents and/or regimens. For examples, methods can further include administering one or more (e.g., two, three, four, five, six, or more) additional agents.
The chemical entity can be administered in combination with one or more additional therapeutic agents and/or regimens that are useful for treating other STING or cGAS-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis. The chemical entity can be administered in combination with one or more additional cancer therapies (e.g., surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof; e.g., chemotherapy that includes administering one or more (e.g., two, three, four, five, six, or more) additional chemotherapeutic agents. Non-limiting examples of additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g.,azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisom erase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab,
Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an anti helminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 - PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL- 10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein
(LAG3), MHC class II - LAG3, 4-1BB-4-1BB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM
- LIGHT, HVEM-BTL A-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48
- CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2,
HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PDl or PD-L1).
The subject can have cancer; e.g., the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies.
Non-limiting examples of cancer include melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma. In certain embodiments, the cancer can be a refractory cancer.
The chemical entity can be administered intratumorally.
The methods can further include identifying the subject.
Other embodiments include those described in the Detailed Description and/or in the claims.
Additional Definitions
To facilitate understanding of the disclosure set forth herein, a number of additional terms are defined below. Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well-known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Each of the patents, applications, published applications, and other publications that are mentioned throughout the specification and the attached appendices are incorporated herein by reference in their entireties.
As used herein, the term“STING” is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous STING molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.
As used herein, the term“cGAS” is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous cGAS molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.
The term“acceptable” with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
“API” refers to an active pharmaceutical ingredient.
The terms“effective amount” or“therapeutically effective amount,” as used herein, refer to a sufficient amount of a chemical entity (e.g., a formula (I) compound or a pharmaceutically acceptable salt and/or hydrate and/or cocrystal thereof) being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an“effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate“effective” amount in any individual case is determined using any suitable technique, such as a dose escalation study.
The term “excipient” or “pharmaceutically acceptable excipient” means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material. In one embodiment, each component is“pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed:, Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 6th ed !; Rowe el al. , Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed !; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical
Preformulation and Formulation, 2nd ed !; Gibson Ed.; CRC Press LLC: Boca Raton, FL, 2009.
The term“pharmaceutically acceptable salt” refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In certain instances, pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. In some instances, pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-m ethy 1 -D-gl ucam i n e, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined. The pharmacologically acceptable salt s not specifically limited as far as it can be used in medicaments. Examples of a salt that the compounds described hereinform with a base include the following: salts thereof with inorganic bases such as sodium, potassium, magnesium, calcium, and aluminum; salts thereof with organic bases such as methylamine, ethylamine and ethanolamine; salts thereof with basic amino acids such as lysine and ornithine; and ammonium salt. The salts may be acid addition salts, which are specifically exemplified by acid addition salts with the following: mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid:organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; acidic amino acids such as aspartic acid and glutamic acid.
The term “pharmaceutical composition” refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.
The term“subject” refers to an animal, including, but not limited to, a primate ( e.g ., human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and“patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human.
The terms“treat,”“treating,” and“treatment,” in the context of treating a disease or disorder, are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof. The“treatment of cancer”, refers to one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, (i) slowing down and (ii) complete growth arrest; (2) reduction in the number of tumor cells; (3) maintaining tumor size; (4) reduction in tumor size; (5) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of tumor cell infiltration into peripheral organs; (6) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of metastasis; (7) enhancement of anti-tumor immune response, which may result in (i) maintaining tumor size, (ii) reducing tumor size, (iii) slowing the growth of a tumor, (iv) reducing, slowing or preventing invasion and/or (8) relief, to some extent, of the severity or number of one or more symptoms associated with the disorder.
The term "halo" refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I).
The term "alkyl" refers to a hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, Ci-io indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. Non-limiting examples include methyl, ethyl, No-propyl, N/V-butyl, «-hexyl.
The term "haloalkyl" refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo.
The term "alkoxy" refers to an -O-alkyl radical (e.g., -OCFE).
The term "alkylene" refers to a divalent alkyl (e.g., -CFh-).
The term "alkenyl" refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds. The alkenyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it.
The term "alkynyl" refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds. The alkynyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it.
The term "aryl" refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like.
The term "cycloalkyl" as used herein includes cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyl may include multiple fused and/or bridged rings. Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butane, bicyclo[2.1.0]pentane, bicyclo[l . l . l]pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like. Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic cycloalkyls include spiro[2.2]pentane, spiro[2.5]octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro[4.5]decane, spiro[3.6]decane, spiro[5.5]undecane, and the like.
The term "cycloalkenyl" as used herein includes partially unsaturated cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkenyl group may be optionally substituted. Examples of cycloalkenyl groups include, without limitation, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. Cycloalkenyl groups may have any degree of saturation provided that none of the rings in the ring system are aromatic; and the cycloalkenyl group is not fully saturated overall. Cycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.
The term“heteroaryl”, as used herein, means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl), and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S. Heteroaryl groups can either be unsubstituted or substituted with one or more substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-<i]pyrimidinyl, pyrrolo[2,3- £]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-£]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridine, pyrazolo[4,3-£]pyridinyl, tetrazolyl, chromane, 2,3-dihydrobenzo[7>][l,4]dioxine, benzo[<i][l,3]dioxole, 2,3- dihydrobenzofuran, tetrahydroquinoline, 2,3-dihydrobenzo[7>][l,4]oxathiine, isoindoline, and others. In some embodiments, the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl.
The term "heterocyclyl" refers to a mon-, bi-, tri-, or polycyclic nonaromatic ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like. Heterocyclyl may include multiple fused and bridged rings. Non-limiting examples of fused/bridged heteorocyclyl includes: 2-azabicyclo[1.1.0]butane, 2-azabicyclo[2.1.0]pentane, 2- azabicyclo[l. l. l]pentane, 3-azabicyclo[3.1.0]hexane, 5-azabicyclo[2.1.1]hexane, 3- azabicyclo[3.2.0]heptane, octahydrocyclopenta[c]pyrrole, 3-azabicyclo[4.1.0]heptane, 7- azabicyclo[2.2.1]heptane, 6-azabicyclo[3.1.1]heptane, 7-azabicyclo[4.2.0]octane, 2- azabicyclo[2.2.2]octane, 3-azabicyclo[3.2.1]octane, 2-oxabicyclo[1.1.0]butane, 2- oxabicyclo[2.1.0]pentane, 2-oxabicyclo[l. l.l]pentane, 3-oxabicyclo[3.1.0]hexane, 5- oxabicyclo[2.1.1]hexane, 3-oxabicyclo[3.2.0]heptane, 3-oxabicyclo[4.1.0]heptane, 7- oxabicyclo[2.2.1]heptane, 6-oxabicyclo[3.1.1]heptane, 7-oxabicyclo[4.2.0]octane, 2- oxabicyclo[2.2.2]octane, 3-oxabicyclo[3.2.1]octane, and the like. Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic heterocyclyls include 2- azaspiro[2.2]pentane, 4-azaspiro[2.5]octane, l-azaspiro[3.5]nonane, 2- azaspiro[3.5]nonane, 7-azaspiro[3.5]nonane, 2-azaspiro[4.4]nonane, 6- azaspiro[2.6]nonane, l,7-diazaspiro[4.5]decane, 7-azaspiro[4.5]decane 2,5- diazaspiro[3 6]decane, 3-azaspiro[5.5]undecane, 2-oxaspiro[2.2]pentane, 4- oxaspiro[2.5]octane, l-oxaspiro[3.5]nonane, 2-oxaspiro[3.5]nonane, 7- oxaspiro[3.5]nonane, 2-oxaspiro[4.4]nonane, 6-oxaspiro[2.6]nonane, 1,7- dioxaspiro[4.5]decane, 2,5-dioxaspiro[3.6]decane, l-oxaspiro[5.5]undecane, 3- oxaspiro[5.5]undecane, 3-oxa-9-azaspiro[5.5]undecane and the like.
In addition, atoms making up the compounds of the present embodiments are intended to include all isotopic forms of such atoms. Isotopes, as used herein, include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include 13C and 14C.
In addition, the compounds generically or specifically disclosed herein are intended to include all tautomeric forms. Thus, by way of example, a compound containing the
encompasses the tautomeric form containing the moiety:
Figure imgf000016_0001
, a pyridinyl or pyrimidinyl moiety that is described to be optionally substituted with hydroxyl encompasses pyridone or pyrimidone tautomeric forms.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features and advantages of the invention will be apparent from the description and drawings, and from the claims.
DETAILED DESCRIPTION
This disclosure features chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit the STING pathway, e.g., inhibit (e.g., antagonize) Stimulator of Interferon Genes (“STING”) or inhibit cyclic GMP-AMP Synthase (“cGAS”). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING pathway activation, e.g., increased (e.g., excessive) STING activation (e.g., STING signaling) or increased (e.g., excessive) cGAS activation (e.g., cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also features compositions containing the same as well as methods of using and making the same.
Formula Compounds
Figure imgf000017_0001
In one aspect, provided herein is a compound of Formula (I):
Figure imgf000017_0002
or a pharmaceutically acceptable salt thereof,
wherein:
W is selected from the group consisting of:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(iv) C(=NH);
(v) S(0)i-2;
(vi) S(0)(NRd);
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F);
Q-A is defined according to (A) or (B) below: ί
Q is NH or N(Rq), wherein Rq is Ci-6 alkyl which is optionally substituted with from 1-2 independently selected Ra; or
Rq and R4, taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each of the heteroatoms is independently selected from N, N(H), O, and S(0)o-2.
A is:
(i) -(YA1)n-YA2, wherein:
• n is 0 or 1;
• YA1 is Ci-6 alkylene, which is optionally substituted with from 1-6 Ra and further optionally substituted with one oxo; and
• YA2 is:
(a) C3-20 cycloalkyl, which is optionally substituted with from 1-
4 Rb,
(b) C6-20 aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-20 ring atoms, wherein from 1- 4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc, or
(d) heterocyclyl including from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb, OR
(ii) Ci-io alkyl, which is optionally substituted with from 1-6 independently selected
Ra, or
Q and A, taken together, form:
*© — , wherein / ? denotes point of attachment to W; and
E is heterocyclyl including from 3-16 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb;
R1 is selected from the group consisting of:
NO2, F, S02R4A, S(0)I-2N(R6A)2, CN, C(=0)R4A, C(0)OR5A, C(0)N(R6A)2, S(0)(NRd)(R4A), S(0)(NH)(R4A), P(0)(0R5A)2, P(0)[N(R6A)2]2, B(OR5A)2 and
P(0)(0R5A)N(R6A)2;
R2 is selected from the group consisting of:
H, halo, cyano, OC(0)R4B, NHC(0)R4B, OR5B, SR5B, NHS02R4B, 0P(0)(0R5B)2, Ci-6 alkyl optionally substituted with 1-2 Ra, and heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc; or R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl which is optionally substituted with from 1-2 Ra,
(iv) Ci-6 alkoxy which is optionally substituted with from 1-2 Ra,
(v) Ci-6 haloalkoxy which is optionally substituted with from 1-2 Ra,
(vi) -NReRf,
(vii) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc,
(viii) C6-io aryl, which is optionally substituted with from 1-2 Rc; or
R3 and R4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci- 3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2;
each of R4A, R4B, R5A and R5B is independently selected from the group consisting of:
(i) H; (ii) Ci-6 alkyl optionally substituted with 1-6 Ra; and
(iii) -(W1)q-W2, wherein:
• q is O or l;
• W1 is Ci-3 alkylene, which is optionally substituted with from 1-6 Ra; and
• W2 is:
(a) C3-10 cycloalkyl, which is optionally substituted with from 1-
4 Rb;
(b) C6-10 aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-10 ring atoms, wherein from 1- 4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc; or
(d) heterocyclyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb; each occurrence of R6Ais independently:
(i) H;
(ii) Ci-10 alkyl which is optionally substituted with 1-6 independently selected Ra;
(iii) (Co-3 alkylene)-C3-io cycloalkyl, which is optionally substituted with from
1-4 Rb,
(iv) (Co-3 alkylene)-C6-io aryl, which is optionally substituted with from 1-4 Rc;
(v) (Co-3 alkylene)-heteroaryl, wherein the heteroaryl includes from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc;
(vi) (Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb; or
(vii) Ci-4 alkoxy; or two occurrences of R6A together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R6), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2; each occurrence of Ra is independently selected from the group consisting of: - OH; -F; -Cl; -Br; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=0)0(Ci-4 alkyl); -C(=0)(Ci-4 alkyl); -C(=0)OH; -CON(R’)(R”); -S(0)I-2(NR’R”); -S(0)i-2(Ci-4 alkyl); cyano, and C3- 6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; each occurrence of Rb is independently selected from the group consisting of: Ci- 10 alkyl optionally substituted with from 1-6 independently selected Ra; Ci-4 haloalkyl; - OH; oxo; -F; -Cl; -Br; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=0)(Ci-4 alkyl); - C(=0)0(Ci-4 alkyl); -C(=0)OH; -C(=0)N(R’)(R”); -S(0)I-2(NR’R”); -S(0)i-2(Ci-4 alkyl); cyano; C6-10 aryl optionally substituted with 1-4 independently selected Ci-4 alkyl; and C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; each occurrence of Rc is independently selected from the group consisting of:
(i) halo; (ii) cyano;
(iii) Ci-io alkyl which is optionally substituted with from 1-6 independently selected
Ra;
(iv) C2-6 alkenyl;
(v) C2-6 alkynyl;
(vi) Ci-4 haloalkyl;
(vii) Ci-4 alkoxy;
(viii) Ci-4 haloalkoxy;
(ix) -(Co-3 alkylene)-C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl;
(x) -(Co-3 alkylene)-C6-io aryl optionally substituted with from 1-4 independently selected Ci-4 alkyl;
(xi) -(Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein the heterocyclyl is optionally substituted with from 1-4 independently selected Ci-4 alkyl;
(xii) -S(0)i-2(Ci-4 alkyl);
(xiii) -NReRf;
(xiv) -OH;
(XV) -S(0)I-2(MCR”);
(xvi) -Ci-4 thioalkoxy;
(xvii) -NO2;
(xviii) -C(=0)(Ci-4 alkyl);
(xix) -C(=0)0(Ci-4 alkyl);
(xx) -C(=0)OH, and
(xxi) -C(=0)N(R’)(R”);
Rd is selected from the group consisting of: Ci-6 alkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); -CON(R’)(R”); -S(0)I-2(NR’R”); - S(0)i-2(Ci-4 alkyl); -OH; and Ci-4 alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; Ci-6 alkyl; Ci-6 haloalkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); - CON(R’)(R”); -S(0)I-2(NR’R”); - S(0)i-2(Ci-4 alkyl); -OH; and CM alkoxy; or Re and Rf together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to Re and Rr), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2; and each occurrence of R’ and R” is independently selected from the group consisting of: H and CM alkyl; or R’ and R” together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R”), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2. In one aspect, compounds of Formula (I), or a pharmaceutically acceptable salt thereof, are featured:
Figure imgf000024_0001
or a pharmaceutically acceptable salt thereof,
wherein: W is selected from the group consisting of:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(iv) C(=NH);
(v) S(0)i-2;
(vi) S(0)(NRd);
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g.,
F);
Q-A is defined according to (A) or (B) below:
(A]
Q is NH or N(Rq), wherein Rq is Ci-6 alkyl which is optionally substituted with from 1-2 independently selected Ra; or
Rq and R4, taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each of the heteroatoms is independently selected from N, N(H), O, and S(0)o-2.
A is:
(i) -(YA1)n-YA2, wherein:
• n is 0 or 1;
• YA1 is Ci-6 alkylene, which is optionally substituted with from 1-6 Ra; and
• YA2 is:
(a) C3-20 cycloalkyl, which is optionally substituted with from 1-4 Rb,
(b) C6-20 aryl, which is optionally substituted with from 1-4 Rc; (c) heteroaryl including from 5-20 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc, or
(d) heterocyclyl including from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1- 4 independently selected Rb,
OR
(ii) Ci-io alkyl, which is optionally substituted with from 1-6 independently selected Ra, or
Q and A, taken together, form: /
N— s , wherein f denotes point of attachment to W; and
E is heterocyclyl including from 3-16 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb;
R1 is selected from the group consisting of:
NO2, F, S02R4A, S(0)I-2N(R6A)2, CN, C(=0)R4A, C(0)0R5A, C(0)N(R6A)2,
S(0)(NRd)(R4A), S(0)(NH)(R4A), P(0)(0R5A)2, P(0)[N(R6A)2]2, B(OR5A)2 and
P(0)(0R5A)N(R6A)2;
R2 is selected from the group consisting of: H, halo, cyano, 0C(0)R4B, NHC(0)R4B, OR5B, SR5B, NHS02R4B, 0P(0)(0R5B)2, Ci-e alkyl optionally substituted with 1-2 Ra, and heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc; or
R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci- 3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl which is optionally substituted with from 1-2 Ra,
(iv) Ci-6 alkoxy which is optionally substituted with from 1-2 Ra,
(v) Ci-6 haloalkoxy which is optionally substituted with from 1-2 Ra,
(vi) -NReRf,
(vii) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc,
(viii) C6-io aryl, which is optionally substituted with from 1-2 Rc; or
R3 and R4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; each of R4A, R4B, R5A and R5B is independently selected from the group consisting of:
(i) H;
(ii) Ci-6 alkyl optionally substituted with 1-6 Ra; and
(iii) -(W4)q-W2, wherein:
• q is O or l;
• W1 is Ci-3 alkylene, which is optionally substituted with from 1-6 Ra; and
• W2 is:
(a) C3-10 cycloalkyl, which is optionally substituted with from 1-4 Rb;
(b) C6-10 aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc; or
(d) heterocyclyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2 and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1- 4 independently selected Rb; each occurrence of R6Ais independently:
(i) H;
(ii) Ci-10 alkyl which is optionally substituted with 1-6 independently selected Ra;
(iii) (Co-3 alkylene)-C3-io cycloalkyl, which is optionally substituted with from 1-4 Rb,
(iv) (Co-3 alkylene)-C6-io aryl, which is optionally substituted with from 1-4 Rc;
(v) (Co-3 alkylene)-heteroaryl, wherein the heteroaryl includes from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc; (vi) (Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb; or
(vii) Ci-4 alkoxy; or two occurrences of R6A together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R6), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2; each occurrence of Ra is independently selected from the group consisting of: -
OH; -F; -Cl; -Br; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=0)0(Ci-4 alkyl); -C(=0)(Ci-4 alkyl); -C(=0)OH; -CON(R’)(R”); -S(0)I-2(NR’R”); -S(0)i-2(Ci-4 alkyl); cyano, and C3- 6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; each occurrence of Rb is independently selected from the group consisting of: Ci-
10 alkyl optionally substituted with from 1-6 independently selected Ra; Ci-4 haloalkyl; - OH; oxo; -F; -Cl; -Br; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=0)(Ci-4 alkyl); - C(=0)0(Ci-4 alkyl); -C(=0)OH; -C(=0)N(R’)(R”); -S(0)I-2(NR’R”); -S(0)i-2(Ci-4 alkyl); cyano; C6-10 aryl optionally substituted with 1-4 independently selected Ci-4 alkyl; and C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; each occurrence of Rc is independently selected from the group consisting of:
(i) halo;
(ii) cyano;
(iii) Ci-10 alkyl which is optionally substituted with from 1-6 independently selected Ra; (iv) C2-6 alkenyl;
(v) C2-6 alkynyl;
(vi) Ci-4 haloalkyl;
(vii) Ci-4 alkoxy;
(viii) Ci-4 haloalkoxy;
(ix) -(Co-3 alkylene)-C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl;
(x) -(Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein the heterocyclyl is optionally substituted with from 1-4 independently selected Ci-4 alkyl;
(xi) -S(0)i-2(Ci-4 alkyl);
(xii) -NReRf;
(xiii) -OH;
(xiv) -S(0)I-2(MCR”);
(xv) -Ci-4 thioalkoxy;
(xvi) -NO2;
(xvii) -C(=0)(Ci-4 alkyl);
(xviii) -C(=0)0(Ci-4 alkyl);
(xix) -C(=0)OH, and
(xx) -C(=0)N(R’)(R”);
Rd is selected from the group consisting of: Ci-6 alkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); -CON(R’)(R”); -S(0)I-2(NR’R”); - S(0)i-2(Ci-4 alkyl); -OH; and Ci- 4 alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; Ci-6 alkyl; Ci-6 haloalkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); - CON(R’)(R”); -S(0)I-2(NR,R”); - S(0)i-2(Ci-4 alkyl); -OH; and CM alkoxy; or Re and Rf together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to Re and R1), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2; and each occurrence of R’ and R” is independently selected from the group consisting of: H and Ci-4 alkyl; or R’ and R” together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C 1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R”), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2.
Embodiments can include any one or more of the features delineated below and/or in the claims.
Variable R\ R4A, R5A, R6A
In some embodiments, R1 is selected from NO2, F, S02R4A, S(0)2N(R6A)2, CN, C(=0)R4A, C(0)0R5A, and C(0)N(R6A)2.
In some embodiments, R1 is selected from NO2, S02R4A, and S(0)2N(R6A)2.
In some embodiments, R1 is S02R4A.
In certain embodiments of foregoing, R4A is: (ii) Ci-6 alkyl optionally substituted with 1-6 Ra; or (iii) -(W4)q-W2 (e.g., q=0; and W2 can be C6-10 aryl).
In certain embodiments (when R4A is: (ii) Ci-6 alkyl optionally substituted with 1- 6 Ra; or (iii) -(W4)q-W2), R4A is Ci-6 alkyl optionally substituted with 1-3 Ra. In certain embodiments (when R4A is Ci-6 alkyl optionally substituted with 1-3 Ra), R4A is unsubstituted Ci-6 alkyl (e.g., C 1-3 alkyl, e.g., methyl). As a non-limiting example of the foregoing, R1 can be S(0)2Me.
In certain embodiments (when R4A is Ci-6 alkyl optionally substituted with 1-3 Ra), R4A is Ci-6 alkyl substituted with from 1-3 Ra (e.g., trifluoromethyl). As a non-limiting example of the foregoing, R1 can be S(0)2CF3.
In some embodiments, R4A is -(W4)q-W2. In certain embodiments, q is 0. In other embodiments, q is 1.
In certain embodiments, W1 is C 1-3 alkylene (e.g., CH2).
In certain embodiments, W2 is C6-10 aryl, which is optionally substituted with from
1-4 Rc.
In certain embodiments of foregoing, W2 is phenyl, which is optionally substituted with from 1-2 Rc (e.g., W2 is unsubstituted phenyl). As a non-limiting example of the foregoing, R1 can be S(0)2Ph.
In certain embodiments, W2 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
In certain embodiments, W2 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
In certain embodiments, W2 is heteroaryl including from 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc. As non-limiting examples of the foregoing, W2 can be pyridinyl optionally substituted with from 1-2 independently selected Rc (e.g., W2 can be pyridin-4-yl or pyridine-2 -yl, e.g., W2 can be unsubstituted pyridin-4-yl or pyridin-2-yl).
In certain embodiments, W2 is heteroaryl including from 5 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
As non-limiting examples of the foregoing, W2 can be thiazolyl optionally substituted with from 1-2 independently selected Rc (e.g., W2 can be thiazol-5-yl optionally substituted with 1 Rc).
As non-limiting examples of any of the foregoing embodiments (when R4A is - (W1)q-W2), R1 can be selected from:
Figure imgf000033_0001
In some embodiments, R1 is NO2.
In some embodiments, R1 is S(0)2N(R6A)2. In certain of these embodiments, R1 is S(0)2NHR6A. In certain of these embodiments, R6A is H or Ci-6 alkyl which is optionally substituted with from 1-6 R6 (e.g., H or unsubstituted Ci-6 alkyl). As non-limiting examples, R6A can be S(0)2NH2, S(0)2NHMe, or S^^NffBu.
In some embodiments, R1 is C(0)N(R6A)2.
In certain embodiments, each of R6A is independently selected from:
(i) H; and
(ii) Ci-6 alkyl optionally substituted with from 1-6 Ra.
In certain embodiments of foregoing, one R6A is H; and the other R6A is Ci-6 alkyl optionally substituted with from 1-6 Ra. As non-limiting examples of the foregoing, R1 can be S(0)2NHMe; or R1 can be C(0)NHMe.
In some embodiments, R1 is CN.
In some embodiments, R1 is F.
In some embodiments, R1 is C(=0)OR5A.
In certain embodiments of foregoing, R5A is selected from (i) H; and (ii) Ci-6 alkyl optionally substituted with from 1-6 Ra (e.g., unsubstituted Ci-6 alkyl, e.g., ethyl or methyl).
In some embodiments, R1 is C(=0)R4A.
In certain embodiments of foregoing, R4A is H.
Variable R2
In some embodiments, R2 is selected from the group consisting of: H, halo, cyano, 0C(0)R4B, NHC(0)R4B, OR5B, and SR5B
In certain embodiments, R2 is selected from the group consisting of: H, halo, cyano, 0C(0)R4B, and SR5B
In some embodiments, R2 is selected from H, halo, and cyano.
In some embodiments, R2 is H.
In some embodiments, R2 is halo.
In certain embodiments, R2 is cyano. In certain embodiments of foregoing when R2 is cyano, R4 is F or C(0)N(R6A)2 (e.g., R1 is F).
In some embodiments, each of R4B and R5B is independently selected from: (ii) Ci- 6 alkyl optionally substituted with 1-6 Ra; and (iii) -(W4)q-W2 (e.g., q is 0; or q is 1).
In certain embodiments of foregoing, each of R4B and R5B is independently (iii) - (W4)q-W2 (e.g., q is 0; or q is 1). In certain embodiments of foregoing, q is 0.
In certain embodiments, W2 is selected from:
(b) C6-io aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
As non-limiting examples of foregoing, W2 can be phenyl, which is optionally
substituted with from
Figure imgf000035_0001
In certain embodiments, W2 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
In certain embodiments, W2 is selected heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
As non-limiting examples of foregoing, W2 can
Figure imgf000035_0002
is unsubstituted Ci-6 alkyl).
In some embodiments (e.g., when phenyl, which is
optionally substituted with from 1-2 Rc (e.g.,
Figure imgf000035_0003
In some embodiments (e.g., when R2 is SR5B), R5B is heteroaryl including from 5- 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently
Figure imgf000036_0001
In some embodiments, R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes:
(a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
In some embodiments, R2 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc, such as R2 is imidazolyl optionally substituted with one Rc.
In some embodiments, R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
In certain embodiments of foregoing, R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring is substituted with from 1-2 oxo. As non-limiting examples of the foregoing, R1 and R2 taken together with the atoms to which each is attached can form a ring selected from the following:
Figure imgf000037_0001
In some embodiments, R1 is S02R4A or S(0)I-2N(R6A)2; and R2 is halo such as chloro. In certain of these embodiments, R4A is Ci-6 alkyl; and each R6A is independently Ci-io alkyl or H. As a non-limiting example, R1 can be S(0)2Me or S(0)2NHMe; and R2 can be chloro.
In some embodiments, R1 is CN or C(=0)R4A; and R2 is halo such as chloro. In certain of these embodiments, R4A is Ci-6 alkyl.
Variables R3, R4, and R5
In some embodiments, each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl which is optionally substituted with from 1-2 Ra,
(iv) Ci-6 alkoxy which is optionally substituted with from 1-2 Ra, and
(v) Ci-6 haloalkoxy which is optionally substituted with from 1-2 Ra.
In some embodiments, each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl,
(iv) Ci-6 alkoxy, and
(v) Ci-6 haloalkoxy. Variable R3
In some embodiments, R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R3 is H.
In certain embodiments, R3 is halo (e.g., F).
In certain embodiments, R3 is Ci-6 alkyl (e.g., methyl).
Variable R4
In some embodiments, R4 is selected from the group consisting of:
(i) H,
(ii) halo, and
(iv) Ci-6 alkoxy (e.g., methoxy).
In certain embodiments, R4 is H.
Variable R5
In some embodiments, R5 is H.
Variables R3 and R4
In some embodiments, R3 and R4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
In certain embodiments, R3 and R4 taken together with the carbon to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
In certain embodiments, R3 and R4 taken together with the carbon to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
As non-limiting examples of the foregoing embodiments, R3 and R4 taken together with the carbon to which each is attached can form a ring selected from the following:
Figure imgf000039_0001
Variable W
In some embodiments, W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F). In certain embodiments, W is C(=0).
In certain embodiments, W is C(=S).
In certain embodiments, W is C(=NRd) (e.g., Rd is Ci-6 alkyl, e.g., methyl).
In certain embodiments, W is C(=C-N02).
In certain embodiments, W is C1-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g., F).
In certain embodiments, W is unsubstituted C1-3 alkylene (e.g., CFh).
In certain embodiments, W is C1-3 alkylene substituted with from 1-4 fluoro (e.g., -C(H)(CF3)-).
Variables Q and A
Embodiments wherein O and A are defined according to (A)
In some embodiments, Q and A are defined according to (A).
In some embodiments, Q is NFL
In some embodiments, A is -(YA1)n-YA2. In certain embodiments, n is 0. In some embodiments, n is 1.
In some embodiments, YA1 is C1-3 alkylene (e.g., YA1 is CFh or CH2CH2).
In some embodiments, YA1 is C1-3 alkylene substituted with oxo, such as CH2C(=0)-.
In some embodiments, YA2 is C6-20 aryl, which is optionally substituted with from
1-4 Rc.
In certain embodiments, YA2 is C6-10 aryl, which is optionally substituted with from
1-3 Rc.
In certain embodiments, YA2 is phenyl, which is optionally substituted with from
1-3 Rc.
In certain embodiments, YA2 is phenyl which is substituted with 1 Rc. As a non-limiting example of the foregoing, YA2 can be phenyl substituted with Rc at the para position.
In certain embodiments, YA2 is C9-10 aryl, which is optionally substituted with from 1-2 Rc.
As a non-limiting example of the foregoing, YA2 can be tetrahydro-naphthyl, which
is optionally substituted with from 1-3 Rc (e.g., YA2 can be
Figure imgf000041_0001
In some embodiments, YA2 is heteroaryl including from 6-10 (e.g., 8-10) ring atoms, wherein from 1-4 (e.g., from 1-2, e.g., 1) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
In certain embodiments, YA2 heteroaryl including from 8-10 ring atoms, wherein from 1-2, such as 1, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
As a non-limiting example of the foregoing, Y^ can be
Figure imgf000041_0002
In certain embodiments, YA2 is heteroaryl including 5-6, such as 6 ring atoms, wherein from 1-2, such as 1, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc. In certain of these embodiments, YA2 is pyridyl which is optionally substituted with from 1-2 independently selected Rc. As a non-limiting example of the foregoing
embodiments, Y^2 can be:
Figure imgf000042_0001
.
In some embodiments, each occurrence of Rc is independently selected from Ci-io alkyl which is optionally substituted with 1-2 independently selected Ra, Ci-4 alkoxy, and C3-6 cycloalkyl.
In certain embodiments, each Ra substituent of Rc is independently selected from hydroxy and Ci-6 alkoxy.
In certain embodiments, each occurrence of Rc is independently selected from butyl (e.g., n-butyl), propyl (e.g., n-propyl), cyclohexyl, 2-hydroxybut-l-yl, ethoxymethyl, methoxy, and ethoxy.
In certain embodiments, each occurrence of Rc is independently C3-6 cycloalkyl such as cyclobutyl and cyclohexyl or C6-10 aryl such as phenyl.
In some embodiments, A is
Figure imgf000042_0002
e.g., Rc is as defined supra or in claims
75-77, e.g., Rc is n-butyl, cyclohexyl, 2-hydroxybut-l-yl, or ethoxymethyl).
In some embodiments, , A A i iss ^— " ( (ee.g g., , ) (e.g·, Rc is as defined supra or in claims 75-77, e.g., Rc is ethoxy or propyl).
In some embodiments, A is
Figure imgf000042_0003
Rc is as defined supra or in claims 75-77, e.g., Rc is ethoxy). In some embodiments, YA2 is C3-20 cycloalkyl, which is optionally substituted with from 1-4 Rb.
In certain embodiments, YA2 is C5-10 cycloalkyl, which is optionally substituted with from 1-4 Rb.
In certain embodiments, wherein YA2 is C6-8 cycloalkyl, which is optionally substituted with from 1-2 Rb.
In some embodiments, each occurrence of Rb is selected from Ci-10 alkyl optionally substituted with from 1-6 independently selected Ra; C i-4 haloalkyl; -OH; oxo; -F; -Cl; - Br; Ci-4 alkoxy; Ci-4 haloalkoxy; and C3-6 cycloalkyl optionally substituted with from 1-4 independently selected C i-4 alkyl.
In certain embodiments, each occurrence of Rb is Ci-10 alkyl optionally substituted with from 1-6 independently selected Ra; Ci-4 haloalkyl; -F; -Cl; -Br; and Ci-4 alkoxy.
In certain embodiments, each occurrence of Rb is Ci-6 alkyl optionally substituted with from 1-3 independently selected Ra; -F; and Ci-4 alkoxy.
As non-limiting examples of the foregoing embodiments, each occurrence of Rb is n-propyl; n-butyl; ethoxymethyl; -F; and Ci-4 alkoxy (e.g., -F, n-propyl, and ethoxymethyl).
In some embodiments, each occurrence of Rb is C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl or C6-10 aryl.
In some embodiments, A is selected from:
Figure imgf000043_0001
In certain embodiments of foregoing, Rb is as defined in claims 84-87 (e.g., Rb is F; or Rb is n-propyl; or Rb is ethoxymethyl).
In some embodiments, Q is N(Rq). In some embodiments, Rq and R4, taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each heteroatom is independently selected from N, N(H), O, and S(0)o-2.
In certain embodiments, Rq and R4, taken together with the atoms connecting them, forms a ring including 5-6 ring atoms, wherein the ring includes (a) from 2-5 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each heteroatom is independently selected from N, N(H), O, and S(0)o-2.
Embodiments wherein O and A are defined according to (B)
In some embodiments, Q and A are defined according to (B).
In some embodiments, E is heterocyclyl including from 3-12 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2 and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-2 independently selected Rb.
In certain embodiments, E is heterocyclyl including from 3-12 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2,and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1 independently selected Rb.
Figure imgf000044_0001
As a non-limiting example of the foregoing, E is: L
As another non-limiting example,
Figure imgf000044_0002
Non-limiting Combinations
[1]
In some embodiments, the compound is of Formula (1-1):
Figure imgf000045_0001
wherein Rcl is as defined for Rc.
In some embodiments of [1], Rcl is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
In certain of these embodiments, Rcl is selected from Ci-io alkyl which is optionally substituted with 1-2 independently selected Ra, Ci-4 alkoxy, and C3-6 cycloalkyl.
In certain embodiments of [1], Rcl is C3-6 cycloalkyl such as cyclobutyl and cyclohexyl or C6-10 aryl such as phenyl.
In certain embodiments when Rcl is Ci-10 alkyl which is optionally substituted with 1-2 independently selected Ra, each each Ra substituent of Rc is independently selected from hydroxy and Ci-6 alkoxy.
As non-limiting examples, Rc can be selected from butyl (e.g., n-butyl), propyl (e.g., n-propyl), cyclohexyl, 2-hydroxybut-l-yl, ethoxymethyl, methoxy, and ethoxy.
As another example, Rc can be CF3.
In some embodiments of [1], wherein W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2; (vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F).
In certain embodiments, W is C(=0).
In some embodiments of [1], R1 is as defined in any one of claims 2-29 (e.g., R1 is NO2).
In certain of these embodiments, R1 is SCbR4A. In certain of these embodiments, R4A is Ci-6 alkyl optionally substituted with 1-6 Ra (e.g., R4A is unsubstituted Ci-6 alkyl such as methyl). For example, R1 can be S(0)2Me.
In certain other embodiments, R1 can be S(0)2N(R6A)2. In certain of these embodiments, each R6A is independently H or Ci-10 alkyl. For example, R1 can be S(0)2NHMe.
In some embodiments of [1], R2 is as defined in any one of claims 30-41.
In certain of these embodiments, R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, and SR5B (e.g., R2 can be selected from H, halo, and cyano).
In certain embodiments of [1], R2 is halo (e.g., chloro).
In certain embodiments of [1], R2 is halo; and R1 is SCbR4A or S(0)2N(R6A)2.
For example, R2 can be chloro; and R1 can be SCbR4A or S(0)2N(R6A)2, wherein R4A is Ci-6 alkyl; and each R6A is independently H or Ci-10 alkyl.
In some embodiments of [1], R1 and R2 are as defined in any one of claims 42-45.
In some embodiments of [1], each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl which is optionally substituted with from 1-2 Ra, (iv) Ci-6 alkoxy which is optionally substituted with from 1-2 Ra, and
(v) Ci-6 haloalkoxy which is optionally substituted with from 1-2 Ra.
In certain of these embodiments of [1], each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl,
(iv) Ci-6 alkoxy, and
(v) Ci-6 haloalkoxy.
In certain embodiments of [1], R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R3 is H.
In certain embodiments of [1], R4 is selected from the group consisting of:
(i) H,
(ii) halo, and
(iv) Ci-6 alkoxy (e.g., methoxy).
In cetain embodiments, R4 is H.
In certain embodiments of [1], each of R3 and R4 is H.
In some embodiments of [1], R3 and R4 is as defined in any one of claims 51-54.
[2]
In some embodiments, the compound is of Formula (1-2):
Figure imgf000048_0001
wherein Rc2 is as defined for Rc.
In some embodiments of [2], Rc2 is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
In some embdoiments of [2], W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e.g·, F).
In certain embodiments, W is C(=0).
In some embodiments of [2], R1 is as defined in any one of claims 2-29 (e.g., R1 is NO2).
In certain of these embodiments, R1 is S02R4A. In certain of these embodiments, R4A is Ci-6 alkyl optionally substituted with 1-6 Ra (e.g., R4A is unsubstituted Ci-6 alkyl such as methyl). For example, R1 can be S(0)2Me.
In certain other embodiments, R1 can be S(0)2N(R6A)2. In certain of these embodiments, each R6A is independently H or Ci-10 alkyl. For example, R1 can be S(0)2NHMe.
In some embodiments of [2], R2 is as defined in any one of claims 30-41. In certain of these embodiments, R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, and SR5B (e.g., R2 can be selected from H, halo, and cyano).
In certain embodiments of [2], R2 is halo (e.g., chloro).
In certain embodiments of [2], R2 is halo; and R1 is S02R4A or S(0)2N(R6A)2.
For example, R2 can be chloro; and R1 can be S02R4A or S(0)2N(R6A)2, wherein R4A is Ci-6 alkyl; and each R6A is independently H or Ci-io alkyl.
In some embodiments of [2], R1 and R2 are as defined in any one of claims 42-45.
In some embodiments of [2], each of R3, R4, and R5 is as defined in any one of claims 46-47.
In certain embodiments of [2], R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R3 is H.
In certain embodiments of [2], R4 is selected from the group consisting of:
(i) H,
(ii) halo, and
(iv) Ci-6 alkoxy (e.g., methoxy).
In cetain embodiments, R4 is H.
In certain embodiments of [2], each of R3and R4 is H.
In some embodiments of [2], R3 and R4 is as defined in any one of claims 51-54. [3]
In some embodiments, the compound is of Formula (I-3a), (I-3b), or (I-3c):
Figure imgf000050_0001
wherein each Rb3 is an independently selected Rb.
In some embodiments of [3], each Rb3 is an independently selected Rb; and wherein said Rb is as defined in any one of claims 84-87.
In some embdoiments of [3], W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F).
In certain embodiments, W is C(=0). In some embodiments of [3], R1 is as defined in any one of claims 2-29 (e.g., R1 is NO2).
In certain of these embodiments, R1 is S02R4A. In certain of these embodiments, R4A is Ci-6 alkyl optionally substituted with 1-6 Ra (e.g., R4A is unsubstituted Ci-6 alkyl such as methyl). For example, R1 can be S(0)2Me.
In certain other embodiments, R1 can be S(0)2N(R6A)2. In certain of these embodiments, each R6A is independently H or Ci-10 alkyl. For example, R1 can be S(0)2NHMe.
In some embodiments of [3], R2 is as defined in any one of claims 30-41.
In certain of these embodiments, R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, and SR5B (e.g., R2 can be selected from H, halo, and cyano).
In certain embodiments of [3], R2 is halo (e.g., chloro).
In certain embodiments of [3], R2 is halo; and R1 is S02R4A or S(0)2N(R6A)2.
For example, R2 can be chloro; and R1 can be S02R4A or S(0)2N(R6A)2, wherein R4A is Ci-6 alkyl; and each R6A is independently H or Ci-10 alkyl.
In some embodiments of [3], R1 and R2 are as defined in any one of claims 42-45.
In some embodiments of [3], each of R3, R4, and R5 is as defined in any one of claims 46-47.
In certain embodiments of [3], R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R3 is H.
In certain embodiments of [3], R4 is selected from the group consisting of:
(i) H, (ii) halo, and
(iv) Ci-6 alkoxy (e.g., methoxy).
In cetain embodiments, R4 is H.
In certain embodiments, each of R3 and R4 is H.
In some embodiments of [3], R3 and R4 is as defined in any one of claims 51-54.
Figure imgf000052_0001
In some embodiments, the compound is of Formula (1-4)
Figure imgf000052_0002
wherein Rc4 is as defined for Rc.
In some embodiments of [4], Rc4 is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
In some embdoiments of [4], W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F).
In certain embodiments, W is C(=0). In some embodiments of [4], R1 is as defined in any one of claims 2-29 (e.g., R1 is NO2).
In certain of these embodiments, R1 is S02R4A. In certain of these embodiments, R4A is Ci-6 alkyl optionally substituted with 1-6 Ra (e.g., R4A is unsubstituted Ci-6 alkyl such as methyl). For example, R1 can be S(0)2Me.
In certain other embodiments, R1 can be S(0)2N(R6A)2. In certain of these embodiments, each R6A is independently H or Ci-10 alkyl. For example, R1 can be S(0)2NHMe.
In some embodiments of [4], R2 is as defined in any one of claims 30-41.
In certain of these embodiments, R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, and SR5B (e.g., R2 can be selected from H, halo, and cyano).
In certain embodiments of [4], R2 is halo (e.g., chloro).
In certain embodiments of [4], R2 is halo; and R1 is S02R4A or S(0)2N(R6A)2.
For example, R2 can be chloro; and R1 can be S02R4A or S(0)2N(R6A)2, wherein R4A is Ci-6 alkyl; and each R6A is independently H or Ci-10 alkyl.
In some embodiments of [4], R1 and R2 are as defined in any one of claims 42-45.
In some embodiments of [4], each of R3, R4, and R5 is as defined in any one of claims 46-47.
In certain embodiments of [4], R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R3 is H.
In certain embodiments of [4], R4 is selected from the group consisting of:
(i) H,
(ii) halo, and (iv) Ci-6 alkoxy (e.g., methoxy).
In cetain embodiments, R4 is H.
In certain embodiments, R5 is H.
In certain embodiments of [4], each of R3 and R4 is H.
In some embodiments of [4], R3 and R4 is as defined in any one of claims 51-54.
[5]
In some embodiments, the compound is of Formula (1-5)
Figure imgf000054_0001
wherein Rc5 is as defined for Rc.
In some embodiments of [5], Rc5 is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
In some embdoiments of [5], W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F).
In certain embodiments, W is C(=0) or CH2. In some embodiments of [5], R1 is as defined in any one of claims 2-29 (e.g., R1 02).
In some embodiments of [5], R2 is as defined in any one of claims 30-41.
In some embodiments of [5], R1 and R2 are as defined in any one of claims 42-45.
In some embodiments of [5], each of R3, and R5 is as defined in any one of claims-47.
In certain embodiments, R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R5 is H.
In certain embodiments of [5], each of R3 and R5 is H.
[6]
In some embodiments, the compound is of Formula (1-6):
Figure imgf000055_0001
In some embdoiments of [6], W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and (ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F).
In certain embodiments, W is C(=0).
In some embdoiments of [6], R1 is as defined in any one of claims 2-29 (e.g., R1 is NO2).
In certain of these embodiments, R1 is S02R4A. In certain of these embodiments, R4A is Ci-6 alkyl optionally substituted with 1-6 Ra (e.g., R4A is unsubstituted Ci-6 alkyl such as methyl). For example, R1 can be S(0)2Me.
In certain other embodiments, R1 can be S(0)2N(R6A)2. In certain of these embodiments, each R6A is independently H or Ci-10 alkyl. For example, R1 can be S(0)2NHMe.
In some embodiments of [6], R2 is as defined in any one of claims 30-41.
In certain of these embodiments, R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, and SR5B (e.g., R2 can be selected from H, halo, and cyano).
In certain embodiments of [6], R2 is halo (e.g., chloro).
In certain embodiments of [6], R2 is halo; and R1 is S02R4A or S(0)2N(R6A)2.
For example, R2 can be chloro; and R1 can be S02R4A or S(0)2N(R6A)2, wherein R4A is Ci-6 alkyl; and each R6A is independently H or Ci-10 alkyl
In some embdoiments of [6], R1 and R2 are as defined in any one of claims 42-45.
In some embdoiments of [6], each of R3, R4, and R5 is as defined in any one of claims 46-47.
In certain embodiments of [6], R3 is selected from the group consisting of:
(i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
In certain embodiments, R3 is H. In certain embodiments of [6], R4 is selected from the group consisting of:
(i) H,
(ii) halo, and
(iv) Ci-6 alkoxy (e.g., methoxy).
In cetain embodiments, R4 is H.
In some embdoiments of [6], R5 is H.
In certain embodiments, each of R3 and R4 is H.
In some embdoiments of [6], R3 and R4 is as defined in any one of claims 51-54.
In some embodiments, the compound is selected from the group of compounds listed in Table 1 :
Table 1.
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
or a pharmaceutically acceptable salt thereof.
This specification concludes with 174 claims. For ease of exposition, certain variable definitions refer to one or more specific claim numbers, and as such, it is understood that the entire subject matter of each claim referenced is incorporated by reference in its entirety into the portion of the disclosure, in which it is referenced. For the avoidance of doubt and as a non-limiting example, use of a phrase, such as“R1 and R2 are as defined in any one of claims 42-45” is intended to represent a short-hand recitation for the following set of defmtions:
R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; OR R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; OR R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring is substituted with from 1-2 oxo; OR R1 and R2 taken together with the atoms to which each is attached forms a ring selected from the following:
Figure imgf000071_0001
Pharmaceutical Compositions and Administration
General
In some embodiments, a chemical entity (e.g., a compound that inhibits (e.g., antagonizes) STING, or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination thereof) is administered as a pharmaceutical composition that includes the chemical entity and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein. In some embodiments, the chemical entities can be administered in combination with one or more conventional pharmaceutical excipients. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, and wool fat. Cyclodextrins such as a-, b, and g-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl-P-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein. Dosage forms or compositions containing a chemical entity as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared. The contemplated compositions may contain 0.001%-100% of a chemical entity provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy , 22nd Edition (Pharmaceutical Press, London, UK. 2012).
Routes of Administration and Composition Components
In some embodiments, the chemical entities described herein or a pharmaceutical composition thereof can be administered to subject in need thereof by any accepted route of administration. Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intracisternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, nasal, nasogastric, oral, parenteral, percutaneous, peridural, rectal, respiratory (inhalation), subcutaneous, sublingual, submucosal, topical, transdermal, transmucosal, transtracheal, ureteral, urethral and vaginal. In certain embodiments, a preferred route of administration is parenteral (e.g., intratumoral).
Compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. The preparation of such formulations will be known to those of skill in the art in light of the present disclosure.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
The carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The 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 dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Intratumoral injections are discussed, e.g., in Lammers, et al., “Effect of
Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems” Neoplasia. 2006, 10, 788-795.
Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository, include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p- oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylocaprate, isopropyl alcohol, propylene glycol, liquid paraffin, xanthan gum, carboxy -metabisulfite, sodium edetate, sodium benzoate, potassium metabi sulfite, grapefruit seed extract, methyl sulfonyl methane (MSM) , lactic acid, glycine, vitamins, such as vitamin A and E and potassium acetate.
In certain embodiments, suppositories can be prepared by mixing the chemical entities described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound. In other embodiments, compositions for rectal administration are in the form of an enema.
In other embodiments, the compounds described herein or a pharmaceutical composition thereof are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.).
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the chemical entity is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
In one embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a chemical entity provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g, in propylene carbonate, vegetable oils, PEG’s, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which one or more chemical entities provided herein or additional active agents are physically separated are also contemplated; e.g. , capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two- compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.
Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid.
In certain embodiments the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient.
In certain embodiments, solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the chemical entity to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel. Exemplary formulation techniques are described in, e.g., Filipski, K.J., et ak, Current Topics in Medicinal Chemistry, 2013, 13, 776-802, which is incorporated herein by reference in its entirety.
Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls.
Other examples include lower-GI targeting techniques. For targeting various regions in the intestinal tract, several enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid-methyl methacrylate copolymers), and Marcoat). Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap.
Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, ETDA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxychloro complex; Allergan, Inc.)).
Topical compositions can include ointments and creams. Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives. Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil. Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase, also sometimes called the“internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and non sensitizing.
In any of the foregoing embodiments, pharmaceutical compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers.
Dosages
The dosages may be varied depending on the requirement of the patient, the severity of the condition being treating and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts. The total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery.
In some embodiments, the compounds described herein are administered at a dosage of from about 0.001 mg/Kg to about 500 mg/Kg (e.g., from about 0.001 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 150 mg/Kg; from about 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 50 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 5 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from about 0.01 mg/Kg to about 0.5 mg/Kg; from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0. 1 mg/Kg to about 200 mg/Kg; from about 0. 1 mg/Kg to about 150 mg/Kg; from about 0. 1 mg/Kg to about 100 mg/Kg; from about 0.1 mg/Kg to about 50 mg/Kg; from about 0. 1 mg/Kg to about 10 mg/Kg; from about 0. 1 mg/Kg to about 5 mg/Kg; from about 0. 1 mg/Kg to about 1 mg/Kg; from about 0. 1 mg/Kg to about 0.5 mg/Kg).
Regimens
The foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month).
In some embodiments, the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 4 months,
5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In an embodiment, a therapeutic compound is administered to an individual for a period of time followed by a separate period of time. In another embodiment, a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped. In an aspect of this embodiment, the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time. In a further embodiment, a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days,
6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
Methods of Treatment
In some embodiments, methods for treating a subject having condition, disease or disorder in which increased (e.g., excessive) STING pathway activity, e.g., increased (e.g., excessive) STING or cGAS activity (e.g., , e.g., STING or cGAS signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., immune disorders, cancer) are provided. Indications
In some embodiments, the condition, disease or disorder is cancer. Non-limiting examples of cancer include melanoma, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include breast cancer, colon cancer, rectal cancer, colorectal cancer, kidney or renal cancer, clear cell cancer lung cancer including small-cell lung cancer, non- small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, squamous cell cancer (e.g. epithelial squamous cell cancer), renal clear cell carcinoma, uveal melanoma, tongue squamous cell carcinoma, breast cancer, skin cancer, cervical cancer, ovarian cancer, prostate cancer, prostatic neoplasms, liver cancer, bladder cancer, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, gastrointestinal stromal tumor, pancreatic cancer, head and neck cancer, glioblastoma, retinoblastoma, astrocytoma, thecomas, arrhenoblastomas, hepatoma, hematologic malignancies including non-Hodgkins lymphoma (NHL), multiple myeloma, myelodysplasia disorders, myeloproliferative disorders, chronic myelogenous leukemia, and acute hematologic malignancies, endometrial or uterine carcinoma, endometriosis, endometrial stromal sarcoma, fibrosarcomas, choriocarcinoma, salivary gland carcinoma, vulval cancer, thyroid cancer, esophageal carcinomas, hepatic carcinoma, anal carcinoma, penile carcinoma, nasopharyngeal carcinoma, laryngeal carcinomas, Kaposi's sarcoma, mast cell sarcoma, ovarian sarcoma, uterine sarcoma, melanoma, malignant mesothelioma, skin carcinomas, Schwannoma, oligodendroglioma, neuroblastomas, neuroectodermal tumor, rhabdomyosarcoma, osteogenic sarcoma, leiomyosarcomas, Ewing Sarcoma, peripheral primitive neuroectodermal tumor, urinary tract carcinomas, thyroid carcinomas, Wilm's tumor, as well as abnormal vascular proliferation associated with phakomatoses, edema (such as that associated with brain tumors), and Meigs' syndrome. In some cases, the cancer is melanoma.
In some embodiments, the condition, disease or disorder is a neurological disorder, which includes disorders that involve the central nervous system (brain, brainstem and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (parts of which are located in both central and peripheral nervous system). Non-limiting examples of cancer include acquired epileptiform aphasia; acute disseminated encephalomyelitis; adrenoleukodystrophy; age-related macular degeneration; agenesis of the corpus callosum; agnosia; Aicardi syndrome; Alexander disease; Alpers' disease; alternating hemiplegia; Alzheimer's disease; Vascular dementia; amyotrophic lateral sclerosis; anencephaly; Angelman syndrome; angiomatosis; anoxia; aphasia; apraxia; arachnoid cysts; arachnoiditis; Anronl-Chiari malformation; arteriovenous malformation; Asperger syndrome; ataxia tel egi ectasia; attention deficit hyperactivity disorder; autism; autonomic dysfunction; back pain; Batten disease; Behcet's disease; Bell's palsy; benign essential blepharospasm; benign focal; amyotrophy; benign intracranial hypertension; Binswanger's disease; blepharospasm; Bloch Sulzberger syndrome; brachial plexus injury; brain abscess; brain injury; brain tumors (including glioblastoma multiforme); spinal tumor; Brown-Sequard syndrome; Canavan disease; carpal tunnel syndrome; causalgia; central pain syndrome; central pontine myelinolysis; cephalic disorder; cerebral aneurysm; cerebral arteriosclerosis; cerebral atrophy; cerebral gigantism; cerebral palsy; Charcot-Marie-Tooth disease; chemotherapy-induced neuropathy and neuropathic pain; Chian malformation; chorea; chronic inflammatory demyelinating polyneuropathy; chronic pain; chronic regional pain syndrome; Coffin Lowry syndrome; coma, including persistent vegetative state; congenital facial diplegia; corticobasal degeneration; cranial arteritis; craniosynostosis; Creutzfeldt-Jakob disease; cumulative trauma disorders; Cushing's syndrome; cytomegalic inclusion body disease; cytomegalovirus infection; dancing eyes-dancing feet syndrome; Dandy-Walker syndrome; Dawson disease; De Morsier's syndrome; Dejerine-Klumke palsy; dementia; dermatomyositis; diabetic neuropathy; diffuse sclerosis; dysautonomia; dysgraphia; dyslexia; dystonias; early infantile epileptic encephalopathy; empty sella syndrome; encephalitis; encephaloceles; encephalotrigeminal angiomatosis; epilepsy; Erb's palsy; essential tremor; Fabry's disease; Fahr's syndrome; fainting; familial spastic paralysis; febrile seizures; Fisher syndrome; Friedreich's ataxia; fronto-temporal dementia and other “tauopathies”; Gaucher's disease; Gerstmann's syndrome; giant cell arteritis; giant cell inclusion disease; globoid cell leukodystrophy; Guillain-Barre syndrome; FITLY- 1- associated myelopathy; Hallervorden-Spatz disease; head injury; headache; hemifacial spasm; hereditary spastic paraplegia; heredopathia atactica polyneuritiformis; herpes zoster oticus; herpes zoster; Hirayama syndrome; HIV-associated dementia and neuropathy (also neurological manifestations of AIDS); holoprosencephaly; Huntington's disease and other polyglutamine repeat diseases; hydranencephaly; hydrocephalus; hypercortisolism; hypoxia; immune-mediated encephalomyelitis; inclusion body myositis; incontinentia pigmenti; infantile phytanic acid storage disease; infantile refsum disease; infantile spasms; inflammatory myopathy; intracranial cyst; intracranial hypertension; Joubert syndrome; Keams-Sayre syndrome; Kennedy disease Kinsbourne syndrome; Klippel Feil syndrome; Krabbe disease; Kugelberg-Welander disease; kuru; Lafora disease; Lambert-Eaton myasthenic syndrome; Landau-Kleffner syndrome; lateral medullary (Wallenberg) syndrome; learning disabilities; Leigh's disease; Lennox-Gustaut syndrome; Lesch-Nyhan syndrome; leukodystrophy; Lewy body dementia; Lissencephaly; locked-in syndrome; Lou Gehrig's disease (i.e., motor neuron disease or amyotrophic lateral sclerosis); lumbar disc disease; Lyme disease— neurological sequelae; Machado-Joseph disease; macrencephaly; megalencephaly; Melkersson-Rosenthal syndrome; Menieres disease; meningitis; Menkes disease; metachromatic leukodystrophy; microcephaly; migraine; Miller Fisher syndrome; mini-strokes; mitochondrial myopathies; Mobius syndrome; monomelic amyotrophy; motor neuron disease; Moyamoya disease; mucopolysaccharidoses; milti-infarct dementia; multifocal motor neuropathy; multiple sclerosis and other demyelinating disorders; multiple system atrophy with postural hypotension; p muscular dystrophy; myasthenia gravis; myelinoclastic diffuse sclerosis; myoclonic encephalopathy of infants; myoclonus; myopathy; myotonia congenital; narcolepsy; neurofibromatosis; neuroleptic malignant syndrome; neurological manifestations of AIDS; neurological sequelae of lupus; neuromyotonia; neuronal ceroid lipofuscinosis; neuronal migration disorders; Niemann-Pick disease; O'Sullivan-McLeod syndrome; occipital neuralgia; occult spinal dysraphism sequence; Ohtahara syndrome; olivopontocerebellar atrophy; opsoclonus myoclonus; optic neuritis; orthostatic hypotension; overuse syndrome; paresthesia; Parkinson's disease; paramyotonia congenital; paraneoplastic diseases; paroxysmal attacks; Parry Romberg syndrome; Pelizaeus-Merzbacher disease; periodic paralyses; peripheral neuropathy; painful neuropathy and neuropathic pain; persistent vegetative state; pervasive developmental disorders; photic sneeze reflex; phytanic acid storage disease; Pick's disease; pinched nerve; pituitary tumors; polymyositis; porencephaly; post-polio syndrome; postherpetic neuralgia; postinfectious encephalomyelitis; postural hypotension; Prader-Willi syndrome; primary lateral sclerosis; prion diseases; progressive hemifacial atrophy; progressive multifocal leukoencephalopathy; progressive sclerosing poliodystrophy; progressive supranuclear palsy; pseudotumor cerebri; Ramsay-Hunt syndrome (types I and II); Rasmussen's encephalitis; reflex sympathetic dystrophy syndrome; Refsum disease; repetitive motion disorders; repetitive stress injuries; restless legs syndrome; retrovirus- associated myelopathy; Rett syndrome; Reye's syndrome; Saint Vitus dance; Sandhoff disease; Schilder's disease; schizencephaly; septo-optic dysplasia; shaken baby syndrome; shingles; Shy-Drager syndrome; Sjogren's syndrome; sleep apnea; Soto's syndrome; spasticity; spina bifida; spinal cord injury; spinal cord tumors; spinal muscular atrophy; Stiff-Person syndrome; stroke; Sturge-Weber syndrome; subacute sclerosing panencephalitis; subcortical arteriosclerotic encephalopathy; Sydenham chorea; syncope; syringomyelia; tardive dyskinesia; Tay-Sachs disease; temporal arteritis; tethered spinal cord syndrome; Thomsen disease; thoracic outlet syndrome; Tic Douloureux; Todd's paralysis; Tourette syndrome; transient ischemic attack; transmissible spongiform encephalopathies; transverse myelitis; traumatic brain injury; tremor; trigeminal neuralgia; tropical spastic paraparesis; tuberous sclerosis; vascular dementia (multi-infarct dementia); vasculitis including temporal arteritis; Von Hippel-Lindau disease; Wallenberg's syndrome; Werdnig-Hoffman disease; West syndrome; whiplash; Williams syndrome; Wildon's disease; amyotrophe lateral sclerosis and Zellweger syndrome.
In some embodiments, the condition, disease or disorder is STING-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis. In certain embodiments, the condition, disease or disorder is an autoimmune disease (e.g., a cytosolic DNA-triggered autoinflammatory disease). Non-limiting examples include rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel diseases (IBDs) comprising Crohn disease (CD) and ulcerative colitis (UC), which are chronic inflammatory conditions with polygenic susceptibility. In certain embodiments, the condition is an inflammatory bowel disease. In certain embodiments, the condition is Crohn’s disease, autoimmune colitis, iatrogenic autoimmune colitis, ulcerative colitis, colitis induced by one or more chemotherapeutic agents, colitis induced by treatment with adoptive cell therapy, colitis associated by one or more alloimmune diseases (such as graft-vs-host disease, e.g., acute graft vs. host disease and chronic graft vs. host disease), radiation enteritis, collagenous colitis, lymphocytic colitis, microscopic colitis, and radiation enteritis. In certain of these embodiments, the condition is alloimmune disease (such as graft-vs-host disease, e.g., acute graft vs. host disease and chronic graft vs. host disease), celiac disease, irritable bowel syndrome, rheumatoid arthritis, lupus, scleroderma, psoriasis, cutaneous T-cell lymphoma, uveitis, and mucositis (e.g., oral mucositis, esophageal mucositis or intestinal mucositis).
In some embodiments, modulation of the immune system by STING provides for the treatment of diseases, including diseases caused by foreign agents. Exemplary infections by foreign agents which may be treated and/or prevented by the method of the present invention include an infection by a bacterium (e.g., a Gram-positive or Gram negative bacterium), an infection by a fungus, an infection by a parasite, and an infection by a virus. In one embodiment of the present invention, the infection is a bacterial infection (e.g., infection by E. coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Salmonella spp., Staphylococcus aureus, Streptococcus spp., or vancomycin-resistant enterococcus), or sepsis. In another embodiment, the infection is a fungal infection (e.g. infection by a mould, a yeast, or a higher fungus). In still another embodiment, the infection is a parasitic infection (e.g., infection by a single-celled or multicellular parasite, including Giardia duodenalis, Cryptosporidium parvum, Cyclospora cayetanensis, and Toxoplasma gondiz). In yet another embodiment, the infection is a viral infection (e.g., infection by a virus associated with AIDS, avian flu, chickenpox, cold sores, common cold, gastroenteritis, glandular fever, influenza, measles, mumps, pharyngitis, pneumonia, rubella, SARS, and lower or upper respiratory tract infection (e.g., respiratory syncytial virus)).
In some embodiments, the condition, disease or disorder is hepatits B (see, e.g., WO 2015/061294).
In some embodiments, the condition, disease or disorder is selected from cardiovascular diseases (including e.g., myocardial infarction).
In some embodiemnts, the condition, disease or disorder is age-related macular degeneration.
In some embodiments, the condition, disease or disorder is mucositis, also known as stomatitits, which can occur as a result of chemotherapy or radiation therapy, either alone or in combination as well as damage caused by exposure to radiation outside of the context of radiation therapy.
In some embodiments, the condition, disease or disorder is uveitis, which is inflammation of the uvea (e.g., anterior uveitis, e.g., iridocyclitis or iritis; intermediate uveitis (also known as pars planitis); posterior uveitis; or chorioretinitis, e.g., pan-uveitis).
In some embodiments, the condition, disease or disorder is selected from the group consisting of a cancer, a neurological disorder, an autoimmune disease, hepatitis B, uvetitis, a cardiovascular disease, age-related macular degeneration, and mucositis.
Still other examples can include those indications discussed herein and below in contemplated combination therapy regimens.
Combination therapy
This disclosure contemplates both monotherapy regimens as well as combination therapy regimens.
In some embodiments, the methods described herein can further include administering one or more additional therapies (e.g., one or more additional therapeutic agents and/or one or more therapeutic regimens) in combination with administration of the compounds described herein. In certain embodiments, the methods described herein can further include administering one or more additional cancer therapies.
The one or more additional cancer therapies can include, without limitation, surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy, cancer vaccines (e.g., HPV vaccine, hepatitis B vaccine, Oncophage, Provenge) and gene therapy, as well as combinations thereof. Immunotherapy, including, without limitation, adoptive cell therapy, the derivation of stem cells and/or dendritic cells, blood transfusions, lavages, and/or other treatments, including, without limitation, freezing a tumor. In some embodiments, the one or more additional cancer therapies is chemotherapy, which can include administering one or more additional chemotherapeutic agents.
In certain embodiments, the additional chemotherapeutic agent is an immunomodulatory moiety, e.g., an immune checkpoint inhibitor. In certain of these embodiments, the immune checkpoint inhibitor targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD-1 - PD-
L2, interleukin-2 (IL-2), indoleamine 2,3 -di oxygenase (IDO), IL-10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR,
CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM - LIGHT, HVEM-BTLA-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48 - CD244,
CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2, HHLA2- TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39-CD73, CXCR4-CXCL 12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155; e.g., CTLA-4 or PDl or PD-Ll). See, e.g., Postow, M. J Clin. Oncol. 2015, 33, 1.
In certain of these embodiments, the immune checkpoint inhibitor is selected from the group consisting of: Urelumab, PF-05082566, MEDI6469, TRX518, Varlilumab, CP-870893, Pembrolizumab (PD1), Nivolumab (PD1), Atezolizumab (formerly
MPDL3280A) (PDL1), MEDI4736 (PD-L1), Avelumab (PD-L1), PDR001 (PD1), BMS-986016, MGA271, Lirilumab, IPH2201, Emactuzumab, INCB024360, Galunisertib,
Ulocuplumab, BKT140, Bavituximab, CC-90002, Bevacizumab, and MNRP1685A, and MGA271.
In certain embodiments, the additional chemotherapeutic agent is an alkylating agent. Alkylating agents are so named because of their ability to alkylate many nucleophilic functional groups under conditions present in cells, including, but not limited to cancer cells. In a further embodiment, an alkylating agent includes, but is not limited to, Cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin. In an embodiment, alkylating agents can function by impairing cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules or they can work by modifying a cell's DNA. In a further embodiment an alkylating agent is a synthetic, semisynthetic or derivative.
In certain embodiments, the additional chemotherapeutic agent is an anti metabolite. Anti-metabolites masquerade as purines or pyrimidines, the building-blocks of DNA and in general, prevent these substances from becoming incorporated in to DNA during the "S" phase (of the cell cycle), stopping normal development and division. Anti- metabolites can also affect RNA synthesis. In an embodiment, an antimetabolite includes, but is not limited to azathioprine and/or mercaptopurine. In a further embodiment an anti metabolite is a synthetic, semisynthetic or derivative. In certain embodiments, the additional chemotherapeutic agent is a plant alkaloid and/or terpenoid. These alkaloids are derived from plants and block cell division by, in general, preventing microtubule function. In an embodiment, a plant alkaloid and/or terpenoid is a vinca alkaloid, a podophyllotoxin and/or a taxane. Vinca alkaloids, in general, bind to specific sites on tubulin, inhibiting the assembly of tubulin into microtubules, generally during the M phase of the cell cycle. In an embodiment, a vinca alkaloid is derived, without limitation, from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea). In an embodiment, a vinca alkaloid includes, without limitation, Vincristine, Vinblastine, Vinorelbine and/or Vindesine. In an embodiment, a taxane includes, but is not limited, to Taxol, Paclitaxel and/or Docetaxel. In a further embodiment a plant alkaloid or terpernoid is a synthetic, semisynthetic or derivative. In a further embodiment, a podophyllotoxin is, without limitation, an etoposide and/or teniposide. In an embodiment, a taxane is, without limitation, docetaxel and/or ortataxel. [021] In an embodiment, a cancer therapeutic is a topoisom erase. Topoisom erases are essential enzymes that maintain the topology of DNA. Inhibition of type I or type II topoisomerases interferes with both transcription and replication of DNA by upsetting proper DNA supercoiling. In a further embodiment, a topoisomerase is, without limitation, a type I topoisomerase inhibitor or a type II topoisomerase inhibitor. In an embodiment a type I topoisomerase inhibitor is, without limitation, a camptothecin. In another embodiment, a camptothecin is, without limitation, exatecan, irinotecan, lurtotecan, topotecan, BNP 1350, CKD 602, DB 67 (AR67) and/or ST 1481. In an embodiment, a type II topoisomerase inhibitor is, without limitation, epipodophyllotoxin. In a further embodiment an epipodophyllotoxin is, without limitation, an amsacrine, etoposid, etoposide phosphate and/or teniposide. In a further embodiment a topoisomerase is a synthetic, semisynthetic or derivative, including those found in nature such as, without limitation, epipodophyllotoxins, substances naturally occurring in the root of American Mayapple (Podophyllum peltatum).
In certain embodiments, the additional chemotherapeutic agent is a stilbenoid. In a further embodiment, a stilbenoid includes, but is not limited to, Resveratrol, Piceatannol, Pinosylvin, Pterostilbene, Alpha- Viniferin, Ampelopsin A, Ampelopsin E, Diptoindonesin C, Diptoindonesin F, Epsilon- Vinferin, Flexuosol A, Gnetin H, Hemsleyanol D, Hopeaphenol, Trans-Diptoindonesin B, Astringin, Piceid and Diptoindonesin A. In a further embodiment a stilbenoid is a synthetic, semisynthetic or derivative.
In certain embodiments, the additional chemotherapeutic agent is a cytotoxic antibiotic. In an embodiment, a cytotoxic antibiotic is, without limitation, an actinomycin, an anthracenedione, an anthracycline, thalidomide, dichloroacetic acid, nicotinic acid, 2- deoxyglucose and/or chlofazimine. In an embodiment, an actinomycin is, without limitation, actinomycin D, bacitracin, colistin (polymyxin E) and/or polymyxin B. In another embodiment, an antracenedione is, without limitation, mitoxantrone and/or pixantrone. In a further embodiment, an anthracycline is, without limitation, bleomycin, doxorubicin (Adriamycin), daunorubicin (daunomycin), epirubicin, idarubicin, mitomycin, plicamycin and/or valrubicin. In a further embodiment a cytotoxic antibiotic is a synthetic, semi sy ntheti c or derivative .
In certain embodiments, the additional chemotherapeutic agent is selected from endostatin, angiogenin, angiostatin, chemokines, angioarrestin, angiostatin (plasminogen fragment), basement-membrane collagen-derived anti-angiogenic factors (tumstatin, canstatin, or arrestin), anti-angiogenic antithrombin III, signal transduction inhibitors, cartilage-derived inhibitor (CDI), CD59 complement fragment, fibronectin fragment, gro- beta, heparinases, heparin hexasaccharide fragment, human chorionic gonadotropin (hCG), interferon alpha/beta/gamma, interferon inducible protein (IP- 10), interleukin- 12, kringle 5 (plasminogen fragment), metalloproteinase inhibitors (TIMPs), 2-methoxyestradiol, placental ribonuclease inhibitor, plasminogen activator inhibitor, platelet factor-4 (PF4), prolactin 16 kD fragment, proliferin-related protein (PRP), various retinoids, tetrahydrocortisol-S, thrombospondin- 1 (TSP-1), transforming growth factor-beta (TGF- b), vasculostatin, vasostatin (calreticulin fragment) and the like. In certain embodiments, the additional chemotherapeutic agent is selected from abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly-l-Lproline-t- butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3',4'-didehydro-4'- deoxy-8'-norvin-caleukoblastine, docetaxol, doxetaxel, cyclophosphamide, carboplatin, carmustine, cisplatin, cryptophycin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, daunorubicin, decitabine dolastatin, doxorubicin (adriamycin), etoposide, 5- fluorouracil, finasteride, flutamide, hydroxyurea and hydroxyureataxanes, ifosfamide, liarozole, lonidamine, lomustine (CCNU), MDV3100, mechlorethamine (nitrogen mustard), melphalan, mivobulin isethionate, rhizoxin, sertenef, streptozocin, mitomycin, methotrexate, taxanes, nilutamide, onapristone, paclitaxel, prednimustine, procarbazine, RPR109881, stramustine phosphate, tamoxifen, tasonermin, taxol, tretinoin, vinblastine, vincristine, vindesine sulfate, and vinflunine.
In certain embodiments, the additional chemotherapeutic agent is platinum, cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, azathioprine, mercaptopurine, vincristine, vinblastine, vinorelbine, vindesine, etoposide and teniposide, paclitaxel, docetaxel, irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide, 5-fluorouracil, leucovorin, methotrexate, gemcitabine, taxane, leucovorin, mitomycin C, tegafur-uracil, idarubicin, fludarabine, mitoxantrone, ifosfamide and doxorubicin. Additional agents include inhibitors of mTOR (mammalian target of rapamycin), including but not limited to rapamycin, everolimus, temsirolimus and deforolimus.
In still other embodiments, the additional chemotherapeutic agent can be selected from those delineated in U.S. Patent 7,927,613, which is incorporated herein by reference in its entirety. In some embodiments, the additional therapeutic agent and/or regimen are those that can be used for treating other STING-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathywith onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis and the like.
Non-limiting examples of additional therapeutic agents and/or regimens for treating rheumatoid arthritis include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), corticosteroids (e.g, prednisone), disease-modifying antirheumatic drugs (DMARDs; e.g., methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), leflunomide (Arava®), hydroxychloroquine (Plaquenil), PF-06650833, iguratimod, tofacitinib (Xeljanz®), ABBV-599, evobrutinib, and sulfasalazine (Azulfidine®)), and biologies (e.g., abatacept (Orencia®), adalimumab (Humira®), anakinra (Kineret®), certolizumab (Cimzia®), etanercept (Enbrel®), golimumab (Simponi®), infliximab (Remicade®), rituximab (Rituxan®), tocilizumab (Actemra®), vobarilizumab, sarilumab (Kevzara®), secukinumab, ABP 501, CHS-0214, ABC-3373, and tocilizumab (ACTEMRA®)).
Non-limiting examples of additional therapeutic agents and/or regimens for treating lupus include steroids, topical immunomodulators (e.g., tacrolimus ointment (Protopic®) and pimecrolimus cream (Elidel®)), thalidomide (Thalomid®), non-steroidal anti inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), antimalarial drugs (e.g., Hydroxychloroquine (Plaquenil)), corticosteroids (e.g, prednisone) and immunomodulators (e.g., evobrutinib, iberdomide, voclosporin, cenerimod, azathioprine (Imuran®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral, Sandimmune®, Gengraf®), and mycophenolate mofetil) baricitinb, iguratimod, filogotinib, GS-9876, rapamycin, and PF-06650833), and biologies (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, obinutuzumab, vobarilizumab, lulizumab, atacicept, PF-06823859, and lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-a-kinoid, OMS721, RC18, RSLY- 132, theralizumab, XmAb5871, and ustekinumab (Stelara®)). For example, non-limiting treatments for systemic lupus erythematosus include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), antimalarial drugs (e.g., Hydroxychloroquine (Plaquenil)), corticosteroids (e.g, prednisone) and immunomodulators (e.g., iberdomide, voclosporin, azathioprine (Imuran®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral, Sandimmune®, Gengraf®), and mycophenolate mofetil, baricitinb, filogotinib, and PF-06650833), and biologies (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDI0700, vobarilizumab, lulizumab, atacicept, PF-06823859, lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-a-kinoid, RC18, RSLV-132, theralizumab, XmAb5871, and ustekinumab (Stelara®)). As another example, non-limiting examples of treatments for cutaneous lupus include steroids, immunomodulators (e.g., tacrolimus ointment (Protopic®) and pimecrolimus cream (Elidel®)), GS-9876, filogotinib, and thalidomide (Thalomid®). Agents and regimens for treating drug-induced and/or neonatal lupus can also be administered.
Non-limiting examples of additional therapeutic agents and/or regimens for treating STING-associated vasculopathy with onset in infancy (SAVI) include JAK inhibitors (e.g., tofacitinib, ruxolitinib, filgotinib, and baricitinib).
Non-limiting examples of additional therapeutic agents and/or regimens for treating Aicardi-Goutieres Syndrome (AGS) include physiotherapy, treatment for respiratory complications, anticonvulsant therapies for seizures, tube-feeding, nucleoside reverse transcriptase inhibitors (e.g., emtricitabine (e.g., Emtriva®), tenofovir (e.g., Viread®), emtricitabine/tenofovir (e.g., Truvada®), zidovudine, lamivudine, and abacavir), and JAK inhibitors (e.g., tofacitinib, ruxolitinib, filgotinib, and baricitinib).
Non-limiting examples of additional therapeutic agents and/or regimens for treating IBDs include 6-mercaptopurine, AbGn-168H, ABX464, ABT-494, adalimumab, AJM300, alicaforsen, AMG139, anrukinzumab, apremilast, ATR-107 (PF0530900), autologous CD34-selected peripheral blood stem cells transplant, azathioprine, bertilimumab, BI 655066, BMS-936557, certolizumab pegol (Cimzia®), cobitolimod, corticosteroids (e.g., prednisone, Methylprednisolone, prednisone), CP-690,550, CT-P13, cyclosporine, DIMS0150, E6007, E6011, etrasimod, etrolizumab, fecal microbial transplantation, figlotinib, fmgolimod, firategrast (SB-683699) (formerly T-0047), GED0301, GLPG0634, GLPG0974, guselkumab, golimumab, GSK 1399686, HMPL-004 ( Andrographis paniculata extract), I1V1U-838, infliximab, Interleukin 2 (IL-2), Janus kinase (JAK) inhibitors, laquinimod, masitinib (AB1010), matrix metalloproteinase 9 (MMP 9) inhibitors (e.g., GS-5745), MEDI2070, mesalamine, methotrexate, mirikizumab (LY3074828), natalizumab, NNC 0142-0000-0002, NNC0114-0006, ozanimod, peficitinib (JNJ-54781532), PF-00547659, PF-04236921, PF-06687234, QAX576, RHB- 104, rifaximin, risankizumab, RPC1063, SB012, SHP647, sulfasalazine, TD-1473, thalidomide, tildrakizumab (MK 3222), TJ301, TNF-Kinoid®, tofacitinib, tralokinumab, TRK-170, upadacitinib, ustekinumab, UTTR1147A, V565, vatelizumab, VB-201, vedolizumab, and vidofludimus.
Non-limiting examples of additional therapeutic agents and/or regimens for treating irritable bowel syndrome include alosetron, bile acid sequesterants (e.g., cholestyramine, colestipol, colesevelam), chloride channel activators (e.g., lubiprostone), coated peppermint oil capsules, desipramine, dicyclomine, ebastine, eluxadoline, famesoid X receptor agonist (e.g., obeticholic acid), fecal microbiota transplantation, fluoxetine, gabapentin, guanylate cyclase-C agonists (e.g., linaclotide, plecanatide), ibodutant, imipramine, JCM- 16021, loperamide, lubiprostone, nortriptyline, ondansetron, opioids, paroxetine, pinaverium, polyethylene glycol, pregabalin, probiotics, ramosetron, rifaximin, and tanpanor.
Non-limiting examples of additional therapeutic agents and/or regimens for treating scleroderma include non-steroidal anti-inflammatory drugs (NSAIDs; e.g., ibuprofen and naproxen), corticosteroids (e.g, prednisone), immunomodulators (e.g., azathioprine, methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral®, Sandimmune®, Gengraf®), antithymocyte globulin, mycophenolate mofetil, intravenous immunoglobulin, rituximab, sirolimus, and alefacept), calcium channel blockers (e.g., nifedipine), alpha blockers, serotonin receptor antagonists, angiotensin II receptor inhibitors, statins, local nitrates, iloprost, phosphodiesterase 5 inhibitors (e.g., sildenafil), bosentan, tetracycline antibiotics, endothelin receptor antagonists, prostanoids, and tyrosine kinase inhibitors (e.g., imatinib, nilotinib and dasatinib).
Non-limiting examples of additional therapeutic agents and/or regimens for treating Crohn’s Disease (CD) include adalimumab, autologous CD34-selected peripheral blood stem cells transplant, 6-mercaptopurine, azathioprine, certolizumab pegol (Cimzia®), corticosteroids (e.g., prednisone), etrolizumab, E6011, fecal microbial transplantation, figlotinib, guselkumab, infliximab, IL-2, JAK inhibitors, matrix metalloproteinase 9 (MMP 9) inhibitors (e.g., GS-5745), MEDI2070, mesalamine, methotrexate, natalizumab, ozanimod, RHB-104, rifaximin, risankizumab, SHP647, sulfasalazine, thalidomide, upadacitinib, V565, and vedolizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating UC include AbGn-168H, ABT-494, ABX464, apremilast, PF-00547659, PF-06687234, 6- mercaptopurine, adalimumab, azathioprine, bertilimumab, brazikumab (MEDI2070), cobitolimod, certolizumab pegol (Cimzia®), CP-690,550, corticosteroids (e.g., multimax budesonide, Methylprednisolone), cyclosporine, E6007, etrasimod, etrolizumab, fecal microbial transplantation, figlotinib, guselkumab, golimumab, IL-2, IMU-838, infliximab, matrix metalloproteinase 9 (MMP9) inhibitors (e.g., GS-5745), mesalamine, mesalamine, mirikizumab (LY3074828), RPC1063, risankizumab (BI 6555066), SHP647, sulfasalazine, TD-1473, TJ301, tildrakizumab (MK 3222), tofacitinib, tofacitinib, ustekinumab, UTTR1147A, and vedolizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating autoimmune colitis include corticosteroids (e.g., budesonide, prednisone, prednisolone, Beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, mesalamine, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating iatrogenic autoimmune colitis include corticosteroids (e.g., budesonide, prednisone, prednisolone, Beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis induced by one or more chemotherapeutics agents include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, mesalamine, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis induced by treatment with adoptive cell therapy include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), diphenoxylate/atropine, infliximab, loperamide, TIP60 inhibitors (see, e.g., U.S. Patent Application Publication No. 2012/0202848), and vedolizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating colitis associated with one or more alloimmune diseases include corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), sulfasalazine, and eicopentaenoic acid.
Non-limiting examples of additional therapeutic agents and/or regimens for treating radaiation enteritis include teduglutide, amifostine, angiotensin-converting enzyme (ACE) inhibitors (e.g., benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, and trandolapril), probiotics, selenium supplementation, statins (e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, and pitavastatin), sucralfate, and vitamin E.
Non-limiting examples of additional therapeutic agents and/or regimens for treating collagenous colitis include 6-mercaptopurine, azathaioprine, bismuth subsalicate, Boswellia serrata extract, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), loperamide, mesalamine, methotrexate, probiotics, and sulfasalazine.
Non-limiting examples of additional therapeutic agents and/or regimens for treating lyphocytic colitis include 6-mercaptopurine, azathioprine, bismuth subsalicylate, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), loperamide, mesalamine, methotrexate, and sulfasalazine.
Non-limiting examples of additional therapeutic agents and/or regimens for treating microscopic colitis include 6-mercaptopurine, azathioprine, bismuth subsalicylate, Boswellia serrata extract, cholestyramine, colestipol, corticosteroids (e.g., budesonide, prednisone, prednisolone, beclometasone dipropionate), fecal microbial transplantation, loperamide, mesalamine, methotrexate, probiotics, and sulfasalazine.
Non-limiting examples of additional therapeutic agents and/or regimens for treating alloimmune disease include intrauterine platelet transfusions, intravenous immunoglobin, maternal steroids, abatacept, alemtuzumab, alphal -antitrypsin, AMG592, antithymocyte globulin, barcitinib, basiliximab, bortezomib, brentuximab, cannabidiol, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, glasdegib, ibrutinib, IL-2, infliximab, itacitinib, LBH589, maraviroc, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, pevonedistat, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.
Non-limiting examples of additional therapeutic agents and/or regimens for treating multiple sclerosis (MS) include alemtuzumab (Lemtrada®), ALKS 8700, amiloride, ATX- MS-1467, azathioprine, baclofen (Lioresal®), beta interferons (e.g., IFN-b- I a, IFN-b- 1 b), cladribine, corticosteroids (e.g., methylprednisolone), daclizumab, dimethyl fumarate (Tecfidera®), fmgolimod (Gilenya®), fluoxetine, glatiramer acetate (Copaxone®), hydroxychloroquine, ibudilast, idebenone, laquinimod, lipoic acid, losartan, masitinib, MD1003 (biotin), mitoxantrone, montelukast, natalizumab (Tysabri®), NeuroVax™, ocrelizumab, ofatumumab, pioglitazone, and RPC 1063. Non-limiting examples of additional therapeutic agents and/or regimens for treating graft-vs-host disease include abatacept, alemtuzumab, alphal -antitrypsin, AMG592, antithymocyte globulin, barcitinib, basiliximab, bortezomib, brentuximab, cannabidiol, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, glasdegib, ibrutinib, IL-2, imatinib, infliximab, itacitinib, LBH589, maraviroc, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, pevonedistat, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.
Non-limiting examples of additional therapeutic agents and/or regimens for treating acute graft-vs-host disease include alemtuzumab, alpha- 1 antitrypsin, antithymocyte globulin, basiliximab, brentuximab, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, defribrotide, denileukin diftitox, ibrutinib, infliximab, itacitinib, LBH589, mycophenolate mofetil, natalizumab, neihulizumab, pentostatin, photopheresis, ruxolitinib, sirolimus, tacrolimus, and tocilizumab.
Non-limiting examples of additional therapeutic agents and/or regimens for treating chronic graft vs. host disease include abatacept, alemtuzumab, AMG592, antithymocyte globulin, basiliximab, bortezomib, corticosteroids (e.g., methylprednisone, prednisone), cyclosporine, dacilzumab, denileukin diftitox, glasdegib, ibrutinib, IL-2, imatinib, infliximab, mycophenolate mofetil, pentostatin, photobiomodulation, photopheresis, ruxolitinib, sirolimus, sonidegib, tacrolimus, tocilizumab, and vismodegib.
Non-limiting examples of additional therapeutic agents and/or regimens for treating celiac disease include AMG 714, AMY01, Aspergillus niger prolyl endoprotease, BL- 7010, CALY-002, GBR 830, Hu-Mik-Beta-1, IMGX003, KumaMax, Larazotide Acetate, Nexvan2®, pancrelipase, TIMP-GLIA, vedolizumab, and ZED1227.
Non-limiting examples of additional therapeutic agents and/or regimens for treating psoriasis include topical corticosteroids, topical crisaborole/AN2728, topical SNA-120, topical SAN021, topical tapinarof, topical tocafmib, topical IDP-118, topical M518101, topical calcipotriene and betamethasone dipropionate (e.g., MC2-01 cream and Taclonex®), topical P-3073, topical LEO 90100 (Enstilar®), topical betamethasone dipropriate (Semivo®), halobetasol propionate (Ultravate®), vitamin D analogues (e.g., calcipotriene (Dovonex®) and calcitriol (Vectical®)), anthralin (e.g., Dritho-scalp® and Dritho-creme®), topical retinoids (e.g., tazarotene (e.g., Tazorac® and Avage®)), calcineurin inhibitors (e.g., tacrolimus (Prograf®) and pimecrolimus (Elidel®)), salicylic acid, coal tar, moisturizers, phototherapy (e.g., exposure to sunlight, UVB phototherapy, narrow band UVB phototherapy, Goeckerman therapy, psoralen plus ultraviolet A (PUVA) therapy, and excimer laser), retinoids (e.g., acitretin (Soriatane®)), methotrexate (Trexall®, Otrexup®, Rasuvo®, Rheumatrex®), Apo805Kl, baricitinib, FP187, KD025, prurisol, VTP-43742, XP23829, ZPL-389, CF101 (piclidenoson), LAS41008, VPD-737 (serlopitant), upadacitinib (ABT-494), aprmilast, tofacitibin, cyclosporine (Neoral®, Sandimmune®, Gengraf®), biologies (e.g., etanercept (Enbrel®), entanercept-szzs (Elrezi®), infliximab (Remicade®), adalimumab (Humira®), adalimumab-adbm (Cyltezo®), ustekinumab (Stelara®), golimumab (Simponi®), apremilast (Otezla®), secukinumab (Cosentyx®), certolixumab pegol, secukinumab, tildrakizumab-asmn, infliximab-dyyb, abatacept, ixekizumab (Taltz®), ABP 710, BCD-057, BI695501, bimekizumab (UCB4940), CHS-1420, GP2017, guselkumab (CNTO 1959), HD203, M923, MSB 11022, Mirikizumab (LY3074828), PF-06410293, PF-06438179, risankizumab (BI655066), SB2, SB4, SB5, siliq (brodalumab), namilumab (MT203, tildrakizumab (MK-3222), and ixekizumab (Taltz®)), thioguanine, and hydroxyurea (e.g., Droxia® and Hydrea®).
Non-limiting examples of additional therapeutic agents and/or regimens for treating cutaneous T-cell lymphoma include phototherapy (e.g., exposure to sunlight, UVB phototherapy, narrow band UVB phototherapy, Goeckerman therapy, psoralen plus ultraviolet A (PUVA) therapy, and excimer laser), extracorporeal photopheresis, radiation therapy (e.g., spot radiation and total skin body electron beam therapy), stem cell transplant, corticosteroids, imiquimod, bexarotene gel, topical bis-chloroethyl-nitrourea, mechlorethamine gel, vorinostat (Zolinza®), romidepsin (Istodax®), pralatrexate (Folotyn®) biologies (e.g., alemtuzumab (Campath®), brentuximab vedotin (SGN-35), mogamulizumab, and IPH4102).
Non-limiting examples of additional therapeutic agents and/or regimens for treating uveitis include corticosteroids (e.g., intravitreal triamcinolone acetonide injectable suspensions), antibiotics, antivirals (e.g., acyclovir), dexamethasone, immunomodulators (e.g., tacrolimus, leflunomide, cyclophosphamide (Cytoxan®, Neosar®, Endoxan®), and cyclosporine (Neoral®, Sandimmune®, Gengraf®), chlorambucil, azathioprine, methotrexate, and mycophenolate mofetil), biologies (e.g., infliximab (Remicade®), adalimumab (Humira®), etanercept (Enbrel®), golimumab (Simponi®), certolizumab (Cimzia®), rituximab (Rituxan®), abatacept (Orencia®), basiliximab (Simulect®), anakinra (Kineret®), canakinumab (Ilaris®), gevokixumab (XOMA052), tocilizumab (Actemra®), alemtuzumab (Campath®), efalizumab (Raptiva®), LFG316, sirolimus (Santen®), abatacept, sarilumab (Kevzara®), and daclizumab (Zenapax®)), cytotoxic drugs, surgical implant (e.g., fluocinolone insert), and vitrectomy.
Non-limiting examples of additional therapeutic agents and/or regimens for treating mucositis include AGO 13, SGX942 (dusquetide), amifostine (Ethyol®), cryotherapy, cepacol lonzenges, capsaicin lozenges, mucoadhesives (e.g., MuGard®) oral diphenhydramine (e.g., Benadry® elixir), oral bioadherents (e.g., polyvinylpyrrolidone- sodium hyaluronate gel (Gelclair®)), oral lubricants (e.g., Oral Balance®), caphosol, chamomilla recutita mouthwash, edible grape plant exosome, antiseptic mouthwash (e.g., chlorhexidine gluconate (e.g., Peridex® or Periogard®), topical pain relievers (e.g., lidocaine, benzocaine, dyclonine hydrochloride, xylocaine (e.g., viscous xylocaine 2%), and Ulcerease® (0.6% phenol)), corticosteroids (e.g., prednisone), pain killers (e.g., ibuprofen, naproxen, acetaminophen, and opioids), GC4419, palifermin (keratinocyte growth factor; Kepivance®), ATL-104, clonidine lauriad, IZN-6N4, SGX942, rebamipide, nepidermin, soluble b-1,3/1,6 glucan, P276, LP-0004-09, CR-3294, ALD-518, IZN-6N4, quercetin, granules comprising vaccinium myrtillus extract, macleaya cordata alkaloids and echinacea angustifolia extract (e.g., SAMITAL®), and gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)). For example, non limiting examples of treatments for oral mucositis include AG013, amifostine (Ethyol®), cryotherapy, cepacol lonzenges, mucoadhesives (e.g., MuGard®) oral diphenhydramine (e.g., Benadry® elixir), oral bioadherents (e.g., polyvinylpyrrolidone-sodium hyaluronate gel (Gelclair®)), oral lubricants (e.g., Oral Balance®), caphosol, chamomilla recutita mouthwash, edible grape plant exosome, antiseptic mouthwash (e.g., chlorhexidine gluconate (e.g., Peridex® or Periogard®), topical pain relievers (e.g., lidocaine, benzocaine, dyclonine hydrochloride, xylocaine (e.g., viscous xylocaine 2%), and Ulcerease® (0.6% phenol)), corticosteroids (e.g., prednisone), pain killers (e.g., ibuprofen, naproxen, acetaminophen, and opioids), GC4419, palifermin (keratinocyte growth factor; Kepivance®), ATL-104, clonidine lauriad, IZN-6N4, SGX942, rebamipide, nepidermin, soluble b-1,3/1,6 glucan, P276, LP-0004-09, CR-3294, ALD-518, IZN-6N4, quercetin, and gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)). As another example, non-limiting examples of treatments for esophageal mucositis include xylocaine (e.g., gel viscous Xylocaine 2%). As another example, treatments for intestinal mucositis, treatments to modify intestinal mucositis, and treatments for intestinal mucositis signs and symptoms include gastrointestinal cocktail (an acid reducer such aluminum hydroxide and magnesium hydroxide (e.g., Maalox), an antifungal (e.g., nystatin), and an analgesic (e.g., hurricane liquid)).
In certain embodiments, the second therapeutic agent or regimen is administered to the subject prior to contacting with or administering the chemical entity (e.g., about one hour prior, or about 6 hours prior, or about 12 hours prior, or about 24 hours prior, or about 48 hours prior, or about 1 week prior, or about 1 month prior).
In other embodiments, the second therapeutic agent or regimen is administered to the subject at about the same time as contacting with or administering the chemical entity. By way of example, the second therapeutic agent or regimen and the chemical entity are provided to the subject simultaneously in the same dosage form. As another example, the second therapeutic agent or regimen and the chemical entity are provided to the subject concurrently in separate dosage forms.
In still other embodiments, the second therapeutic agent or regimen is administered to the subject after contacting with or administering the chemical entity (e.g., about one hour after, or about 6 hours after, or about 12 hours after, or about 24 hours after, or about 48 hours after, or about 1 week after, or about 1 month after).
Patient Selection
In some embodiments, the methods described herein further include the step of identifying a subject (e.g., a patient) in need of such treatment (e.g., by way of biopsy, endoscopy, or other conventional method known in the art). In certain embodiments, the STING protein can serve as a biomarker for certain types of cancer, e.g., colon cancer and prostate cancer. In other embodiments, identifying a subject can include assaying the patient’s tumor microenvironment for the absence of T-cells and/or presence of exhausted T-cells, e.g., patients having one or more cold tumors. Such patients can include those that are resistant to treatment with checkpoint inhibitors. In certain embodiments, such patients can be treated with a chemical entity herein, e.g., to recruit T-cells into the tumor, and in some cases, further treated with one or more checkpoint inhibitors, e.g., once the T-cells become exhausted.
In some embodiments, the chemical entities, methods, and compositions described herein can be administered to certain treatment-resistant patient populations (e.g., patients resistant to checkpoint inhibitors; e.g., patients having one or more cold tumors, e.g., tumors lacking T-cells or exhausted T-cells).
Compound Preparation
As can be appreciated by the skilled artisan, methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and RGM. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. The starting materials used in preparing the compounds of the invention are known, made by known methods, or are commercially available. The skilled artisan will also recognize that conditions and reagents described herein that can be interchanged with alternative art-recognized equivalents. For example, in many reactions, triethylamine can be interchanged with other bases, such as non- nucleophilic bases (e.g. diisopropylamine, l,8-diazabicycloundec-7-ene, 2,6-di-tert- butylpyridine, or tetrabutylphosphazene).
The skilled artisan will recognize a variety of analytical methods that can be used to characterize the compounds described herein, including, for example, ¾ NMR, heteronuclear NMR, mass spectrometry, liquid chromatography, and infrared spectroscopy. The foregoing list is a subset of characterization methods available to a skilled artisan and is not intended to be limiting.
To further illustrate the foregoing, the following non-limiting, exemplary synthetic schemes are included. Variations of these examples within the scope of the claims are within the purview of one skilled in the art and are considered to fall within the scope of the invention as described, and claimed herein. The reader will recognize that the skilled artisan, provided with the present disclosure, and skill in the art is able to prepare and use the invention without exhaustive examples.
Examples
Example 1
The following examples are synthesized from commercial carboxylic acids by the following methods.
Figure imgf000103_0001
1 -1 1 -2 Ex. 1
Amine (e.g., 1-2) (1.0 eq.) and carboxylic acid (e.g., 1-1) (1.0 eq.) are dissolved in 2 ml DMF. 5.0 eq. of triethylamine and 1.0 Eq. of EDC is added. The resulting mixture is stirred for 24h, after which it is poured onto a mixture of DCM and 10% citric acid (1 : 1). The phases are separated, and the aqueous phase is extracted with DCM. The combined organic phases are washed with 10 mL water, dried over MgSCE, and concentrated under vacuum. The solid is dissolved in DCM and adsorbed on 1.2g Silica, followed by flash chromatography (12 g S1O2, elution with hexane/ AcOEt) to yield the purified compound.
Figure imgf000103_0002
1-3 1 -2 Ex. 1
Alternatively the amine (e.g., 1-1) and acid chloride (e.g. 1-3) (prepared from the corresponding acid) are dissolved in 2 mL DMF. 5.0 eq. of triethylamine and 1.0 Eq. of EDC is added and the mixture stirred for 24h. The mixture is poured onto a mixture of DCM and 10% citric acid (1 : 1). The phases are separated, and the aqueous phase is extracted with DCM. The combined organic phases are washed with 10 ml water, dried over MgSCri, and concentrated under vacuum. The solid is dissolved in DCM and adsorbed on 1.2 g silica, followed by flash chromatography: (12 g S1O2, elution with Hexane/ AcOEt) to yield the purified compound. The following examples are synthesized by the method described above from the corresponding acid and amine:
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
The following abbreviations have the indicated meanings:
ACN = acetonitrile
AcOH = acetic acid
DCM = dichloromethane
COCh = Phosgene
Cul = cuprous Iodide
DMF = N,N-dimethylformamide
DMSO = dimethyl sulfoxide
DIEA = N,N-diisopropylethylamine
DPPA = diphenyl azidophosphate
DCE = dichloroethane T3P = 1-propanephosphonic acid cyclic anhydride
H2O = water
HATU = 2-(7-azabenzotriazol-l-yl)-N,N,N,,N’-tetramethyluronium hexafluorophosphate
TBS = tertbutylaimethylsilyl chloride
Py = pyridine
Et = ethyl
EtOAc = ethyl acetate
EtOH = ethanol
LC-MS = liquid chromatography - mass spectrometry
Me = methyl
MeOH = methanol
NaOH = sodium hydroxide
NEE = ammonia
n-Bu = n-butyl
NMR = nuclear magnetic resonance
Ph = phenyl
HPLC = high performance liquid chromatography
t-Bu = tert-butyl
TEA = triethylamine
TFA = trifluoroacetic acid
THF = tetrahydrofuran
TLC = thin layer chromatography Materials and Methods
The progress of reactions was often monitored by TLC or LC-MS. The identity of the products was often confirmed by LC-MS. The LC-MS was recorded using one of the following methods.
Method A: Titank C18, 50x3 mm, 3 um column, 0.3 uL injection, 1.5 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile phase A: Water+5mMNH4HC03 and Mobile Phase B: Acetonitrile. 10% MPB to 95.0% in 1.39 min, hold at 95% MPB for 0.8 min, 95% MPB to 10% in 0.03 min, then equilibration to 10% MPB for 0.27 min. Method B: EVO-C18, 50x3 mm, 2.6 um column, 2.0 uL injection, 1.2 mL/min flow rate, 90-900 amu scan range, 254 nm UV detection. Mobile phase A: Water+5mMNH4HC03 and Mobile Phase B: Acetonitrile. 10% MPB to 95.0% in 1.99 min, hold at 95% MPB for 0.6 min, 95% MPB to 10% in 0.20 min, then equilibration to 10% MPB for 0.25 min. Method D: Poroshell HPH-C18, 50 *3mm, 2.7 pL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.04% NLLOH and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 1.99 min, hold at 95% MPB for 0.6 min, 95% MPB to 10% in 0.03 min, then equilibration to 10% MPB for 0.17 min.
Method L: Shim-pack XR-ODS, 50 *3.0 mm, 50 *3.0 mm, 2.2 um column, 2.2 pL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.05% TFA and Mobile Phase B (MPB): Acetonitrile. Elution 5% MPB to 95% in 2 min, hold at 95% MPB for 0.7 min, 95% MPB to 10% in 0.05 min, then equilibration to 5% MPB for 0.25 min.
The final targets were purified by Prep-HPLC. The Prep-HPLC was carried out using the following method.
Method I: Pre-HPLC: Column: Xselect CSH OBD Column 30* 150mm 5um; Mobile Phase: Water (10MMOL/L NH HCC>3+0.1%NH3.H2O) and ACN, UV detection 254/210 nm.
Method K: Pre-HPLC: Column: Atlantis HILIC OBD Column, 19* 150mm*5um; Mobile Phase: Water(10MMOL/L MLHCOs+O. U/oML.HiO) and ACN, UV detection 254/210 nm.
NMR was recorded on BRUKER NMR 300.03 Mz, DUL-C-H, ULTRASHIELD™ 300, AVANCE II 300 B-ACS™ 120 or BRUKER NMR 400.13 Mz, BBFO, ULTRASHIELD™ 400, AVANCE III 400, B-ACS™ 120.
Preparative examples
Scheme for the preparation of Intermediates:
Scheme 1: Synthesis of intermediate 2 (3-carbamoyl-4-chlorobenzoic acid)
Figure imgf000116_0001
Step 1: Synthesis of methyl 3-carbamoyl-4-chlorobenzoate
Figure imgf000116_0002
2-chloro-5-(methoxycarbonyl)benzoic acid (200 mg, 0.9 mmol, 1.0 equiv) was dissolved in DCM (10 mL). (COCl)2 (0.24 mL, 2.8 mmol, 3.0 equiv) and DMF (cat.) were added. The resulting mixture was stirred for 30 min and was concentrated. The residue thus obtained was dissolved in THF (5.0 mL). To the above, ML (g) in THF (0.5M, 5.0 mL) was added. The resulting solution was stirred for 30 min at ambient temperature. The resulting mixture was concentrated in vacuo. The residue was purified by flash column chromatography on silica gel, eluting with EtOAc/petroleum ether (1 :2) to give methyl 3- carbamoyl-4-chlorobenzoate (160 mg, 80.37%) as a white solid. LCMS: Method A, MS- ESI: 212.0 [M-H] .
Step 2: Synthesis of 3-carbamoyl-4-chlorobenzoic acid
Figure imgf000117_0001
A3 Intermediate 2
Methyl 3 -carbarn oyl-4-chlorobenzoate (200 mg, 0.9 mmol, 1.0 equiv) was dissolved in THF (10.0 mL). EhO (2.0 mL) and NaOH (149.8 mg, 3.7 mmol, 4.0 equiv) were added. The resulting solution was stirred for 6 hours at ambient temperature. The pH value of the solution was adjusted to 2 with HC1 aqueous (6 mol/L). The solids were collected by filtration. 3-carbamoyl-4-chlorobenzoic acid (80 mg, 42.8%) was obtained as a white solid. LCMS: Method A, 198.0 [M-H] .
Scheme 2: Synthesis of intermediate 3 (3-chloro-5-(methylsulfonyl)benzoic acid)
Figure imgf000117_0002
Figure imgf000117_0003
Intermediate 3 Methyl 3-chloro-5-iodobenzoate (500.0 mg, 1.7 mmol, 1.0 equiv) was dissolved in DMSO (10.0 mL). Sodium methanesulfmate (206.6 mg, 2.0 mmol, 1.2 equiv), L-proline (29.1 mg, 0.3 mmol, 0.15 equiv), NaOH (13.5 mg, 0.3 mmol, 0.2 equiv), and Cul (32.1 mg, 0.2 mmol, 0.1 equiv) were added under nitrogen. The resulting solution was stirred for 6 hours at 90 °C and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel, eluting with EtOAc/petroleum ether (1 : 1) to give 3-chloro- 5-methanesulfonylbenzoic acid (148 mg, 37.40%) as brown yellow oil. LCMS: Method A, MS-ESI: 233.0 [M-H] .
Scheme 3: Synthesis of intermediate 4 5-butylpyridin-2-amine hydrochloride
Figure imgf000118_0001
S ep 3 Intermediate 4
Step 1: Synthesis of 5-butylpicolinoyl azide
Figure imgf000118_0002
5-Butylpicolinic acid (800.0 mg, 4.5 mmol, 1.0 equiv.) and DPPA (1.8 g, 6.7 mmol, 1.5 equiv.) were dissolved in THF (10.0 mL). TEA (0.94 mL, 6.7 mmol, 1.5 equiv) was added. Upon stirring 1.5 days at ambient temperature, the resulting mixture was concentrated in vacuo. The crude resulting mixture was used directly in the next step without further purification. LCMS: Method D, 205.1 [M+H]+.
Step 2: Synthesis of tert-butyl (5-butylpyridin-2-yl)carbamate
Figure imgf000119_0001
5-butylpicolinoyl azide (800.0 mg, 3.9 mmol, 1.0 equiv.) was dissolved in t-BuOH
(10.0 mL). Upon stirring 10 hours at 90 °C under nitrogen atmosphere, the resulting mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with petroleum ether /EtOAc (2: 1) to afford tert-butyl N-(5- butylpyridin-2-yl) carbamate (500 mg, 50.9%) as a yellow solid. LCMS: Method D, 251.2 [M+H]+.
Step 3: Synthesis of 5-butylpyridin-2-amine hydrochloride
Figure imgf000119_0002
A7 Intermediate 4
Tert-butyl (5-butylpyridin-2-yl)carbamate (500.0 mg, 2.0 mmol, 1.0 equiv.) was dissolved in HC1 in dioxane (4 M, 5.0 mL). The reaction mixture was stirred for 3 hours at ambient temperature and concentrated in vacuo. 5-Butylpyridin-2-amine hydrochloride (crude) was isolated as a yellow solid. LCMS: Method D, 151.1 [M+H-HC1]+.
Scheme for preparation of Example 71
A: /V-(4-butylphenyl)-4-fluoro-3-nitrobenzamide (Example 71)
Figure imgf000119_0003
Example 71
Figure imgf000120_0001
4-Butylaniline (150.0 mg, 1.0 mmol, 1.0 equiv) was dissolved in THF (5.0 mL). 4- fluoro-3-nitrobenzoic acid (186.2 mg, 1.0 mmol, 1.0 equiv), T3P (670.0 mg, 2.0 mmol, 2.0 equiv) and TEA (0.42 mL, 3.0 mmol, 3.0 equiv) were added. The resulting solution was stirred for 3 hours at ambient temperature and then diluted with 60 mL of EtOAc. The organic layer was concentrated in vacuo. The residue was purified by Prep-HPLC using Method I, to give /V-(4-butylphenyl)-4-fluoro-3-nitrobenzamide (189.7 mg, 59.7%) as an off-white solid. LCMS: Method A, MS-ESI: 317.2 [M+H] +. 1HNMR (300 MHz, DMSO-rfi): d 10.49 (s, 1H), 8.74 (dd, 1H), 8.40-8.37 (m, 1H), 7.80-7.73 (m, 1H), 7.65 (d, 2H), 7.20 (d, 2H), 2.59-2.51 (m, 2H), 1.58-1.53 (m, 2H), 1.34-1.27 (m, 2H),
0.90 (t, 3H).
The following compounds in Table El were synthesized using similar methods as described above for Example 71.
Table El
Figure imgf000120_0002
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
The analogs in Table E2 were prepared using similar methods as described elsewhere herein using the starting materials as described.
Table E2
Figure imgf000123_0002
Figure imgf000124_0001
Biological Assays
STING pathway modulation by the compounds described herein was measured using THPl-Dual™ cells (KO-IFNAR2).
THPl-Dual™ KO-IFNAR2 Cells (obtained from invivogen) were maintained in RPMI, 10% FCS, 5 ml P/S, 2mM L-glut, lOmM Hepes, and 1 mM sodium pyruvate. Compounds were spotted in empty 384 well tissue culture plates (Greiner 781182) by Echo for a final concentration of 0.0017 - 100 mM Cells were plated into the TC plates at 40 pL per well, 2>< 10E6 cells/mL. For activation with STING ligand, 2'3'cGAMP (MW 718.38, obtained from Invivogen), was prepared in Optimem media.
The following solutions were prepared for each 1 ><384 plate:
o Solution A: 2 mL Optimem with one of the following stimuli:
60 uL of 10 mM 2'3'cGAMP -> 150 mM stock
o Solution B: 2 mL Optimem with 60 pL Lipofectamine 2000 -> Incubate 5 min at RT
2 mL of solution A and 2 ml Solution B was mixed and incubated for 20 min at room temperature (RT). 20 pL of transfection solution (A+B) was added on top of the plated cells, with a final 2’3’cGAMP concentration of 15 pM. The plates were then centrifuged immediately at 340 g for 1 minute, after which they were incubated at 37 °C, 5% CO2, >98% humidity for 24h. Luciferase reporter activity was then measured. EC50 values were calculated by using standard methods known in the art.
Luciferase reporter assay: 10 pL of supernatant from the assay was transferred to white 384-plate with flat bottom and squared wells. One pouch of QUANTI-Luc™ Plus was dissolved in 25 mL of water. 100 pL of QLC Stabilizer per 25 mL of QUANTI- Luc™ Plus solution was added. 50 pL of QUANTI-Luc™ Plus/QLC solution per well was then added. Luminescence was measured on a Platereader (e.g., Spectramax I3X (Molecular Devices GF3637001)).
Luciferase reporter activity was then measured. ECso values were calculated by using standard methods known in the art.
Table BA shows the activity of compounds in STING reporter assay: <0.008 mM =
“++++++”; >0.008 and <0.04 pM =“+++++”; >0.04 and <0.2 pM =“++++”; >0.2 and <1 pM =“+++”; >1 and <5 pM =“++”; >5 and <100 pM =“+”.
Table BA
Figure imgf000125_0001
Figure imgf000126_0001

Claims

WHAT IS CLAIMED IS:
1. A method for inhibiting STING or cGAS activity, the method comprising contacting STING or cGAS with a compound of Formula (I):
Figure imgf000127_0001
or a pharmaceutically acceptable salt thereof,
wherein:
W is selected from the group consisting of:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(iv) C(=NH);
(v) S(0)i-2;
(vi) S(0)(NRd);
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g.,
Q-A is defined according to (A) or (B) below:
Figure imgf000127_0002
Q is NH or N(Rq), wherein Rq is Ci-6 alkyl which is optionally substituted with from 1-2 independently selected Ra; or
Rq and R4, taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each of the heteroatoms is independently selected from N, N(H), O, and S(0)o-2.
A is:
(i) -(YA1)n-YA2, wherein:
• n is 0 or 1;
• YA1 is Ci-6 alkylene, which is optionally substituted with from 1-6 Ra and further optionally substituted with one oxo; and
• YA2 is:
(a) C3-20 cycloalkyl, which is optionally substituted with from 1-4 Rb,
(b) C6-20 aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-20 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc, or
(d) heterocyclyl including from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1- 4 independently selected Rb,
OR
(ii) Ci-io alkyl, which is optionally substituted with from 1-6 independently selected Ra, or
Figure imgf000128_0001
Q and A, taken together, form:
- - , wherein f f denotes point of attachment to W; and
E is heterocyclyl including from 3-16 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb;
R1 is selected from the group consisting of:
NO2, F, S02R4A, S(0)I-2N(R6A)2, CN, C(=0)R4A, C(0)0R5A, C(0)N(R6A)2,
S(0)(NRd)(R4A), S(0)(NH)(R4A), P(0)(0R5A)2, P(0)[N(R6A)2]2, B(OR5A)2 and
P(0)(0R5A)N(R6A)2;
R2 is selected from the group consisting of:
H, halo, cyano, 0C(0)R4B, NHC(0)R4B, OR5B, SR5B, NHS02R4B, 0P(0)(0R5B)2, Ci-e alkyl optionally substituted with 1-2 Ra, and heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc; or
R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, Ci- 3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H, (ii) halo,
(iii) Ci-6 alkyl which is optionally substituted with from 1-2 Ra,
(iv) Ci-6 alkoxy which is optionally substituted with from 1-2 Ra,
(v) Ci-6 haloalkoxy which is optionally substituted with from 1-2 Ra,
(vi) -NReRf,
(vii) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc,
(viii) C6-io aryl, which is optionally substituted with from 1-2 Rc; or
R3 and R4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2; each of R4A, R4B, R5A and R5B is independently selected from the group consisting of:
(i) H;
(ii) Ci-6 alkyl optionally substituted with 1-6 Ra; and
(iii) -(W4)q-W2, wherein:
• q is O or l;
• W1 is Ci-3 alkylene, which is optionally substituted with from 1-6 Ra; and
• W2 is:
(a) C3-10 cycloalkyl, which is optionally substituted with from 1-4 Rb;
(b) C6-10 aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc; or
(d) heterocyclyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1- 4 independently selected Rb; each occurrence of R6Ais independently:
(i) H;
(ii) Ci-io alkyl which is optionally substituted with 1-6 independently selected Ra;
(iii) (Co-3 alkylene)-C3-io cycloalkyl, which is optionally substituted with from 1-4 Rb,
(iv) (Co-3 alkylene)-C6-io aryl, which is optionally substituted with from 1-4 Rc;
(v) (Co-3 alkylene)-heteroaryl, wherein the heteroaryl includes from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc;
(vi) (Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-4 independently selected Rb; or
(vii) Ci-4 alkoxy; or two occurrences of R6A together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R6), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2; each occurrence of Ra is independently selected from the group consisting of: - OH; -F; -Cl; -Br; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=0)0(Ci-4 alkyl); -C(=0)(Ci-4 alkyl); -C(=0)0H; -CON(R’)(R”); -S(0)i.2(NR’R”); -S(0)i-2(Ci-4 alkyl); cyano, and C3- 6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; each occurrence of Rb is independently selected from the group consisting of: Ci- lo alkyl optionally substituted with from 1-6 independently selected Ra; Ci-4 haloalkyl; - OH; oxo; -F; -Cl; -Br; -NReRf; Ci-4 alkoxy; Ci-4 haloalkoxy; -C(=0)(Ci-4 alkyl); - C(=0)0(Ci-4 alkyl); -C(=0)OH; -C(=0)N(R’)(R”); -S(0)I-2(NR’R”); -S(0)i-2(Ci-4 alkyl); cyano; C6-10 aryl optionally substituted with 1-4 independently selected Ci-4 alkyl; and C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; each occurrence of Rc is independently selected from the group consisting of:
(i) halo;
(ii) cyano;
(iii) Ci-10 alkyl which is optionally substituted with from 1-6 independently selected Ra;
(iv) C2-6 alkenyl;
(v) C2-6 alkynyl;
(vi) Ci-4 haloalkyl;
(vii) Ci-4 alkoxy;
(viii) Ci-4 haloalkoxy;
(ix) -(Co-3 alkylene)-C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl;
(x) -(Co-3 alkylene)-C6-io aryl optionally substituted with from 1-4 independently selected
Ci-4 alkyl;
(xi) -(Co-3 alkylene)-heterocyclyl, wherein the heterocyclyl includes from 3-16 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein the heterocyclyl is optionally substituted with from 1-4 independently selected Ci-4 alkyl; (xii) -S(0)i-2(Ci-4 alkyl);
(xiii) -NReRf;
(xiv) -OH;
(xv) -S(0)I-2(NR’R”);
(xvi) -Ci-4 thioalkoxy;
(xvii) -NO2;
(xviii) -C(=0)(Ci-4 alkyl);
(xix) -C(=0)0(Ci-4 alkyl);
(xx) -C(=0)OH, and
(xxi) -C(=0)N(R’)(R”);
Rd is selected from the group consisting of: Ci-6 alkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); -CON(R’)(R”); -S(0)I-2(NR’R”); - S(0)i-2(Ci-4 alkyl); -OH; and Ci- 4 alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; Ci-6 alkyl; Ci-6 haloalkyl; C3-6 cycloalkyl; -C(0)(Ci-4 alkyl); -C(0)0(Ci-4 alkyl); - CON(R’)(R”); -S(0)I-2(NR,R”); - S(0)I-2(CM alkyl); -OH; and CM alkoxy; or Re and Rf together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to Re and R1), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2; and each occurrence of R’ and R” is independently selected from the group consisting of: H and Ci-4 alkyl; or R’ and R” together with the nitrogen atom to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 1-7 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H and C1-3 alkyl; and (b) from 0-3 ring heteroatoms (in addition to the nitrogen atom attached to R’ and R”), which are each independently selected from the group consisting of N(H), N(Rd), O, and S(0)o-2.
2. The method of claim 1, wherein R1 is selected from NO2, F, S02R4A, S(0)2N(R6A)2, CN, C(=0)R4A, C(0)0R5A, and C(0)N(R6A)2.
3. The method of any one of claims 1-2, wherein R1 is selected from NO2, S02R4A, and S(0)2N(R6A)2.
4. The method of any one of claims 1-3, wherein R1 is S02R4A.
5. The method of claim 4, wherein R4A is:
(ii) Ci-6 alkyl optionally substituted with 1-6 Ra; or
(iii) -(W4)q-W2 (e.g., q=0; and W2 is C6-10 aryl).
6. The method of claim 5, wherein R4A is Ci-6 alkyl optionally substituted with 1-3 Ra.
7. The method of claim 6, wherein R4A is unsubstituted Ci-6 alkyl (e.g., C1-3 alkyl, e.g., methyl).
8. The method of claim 7, wherein R1 is S(0)2Me.
9. The method of claim 6, wherein R4A is Ci-6 alkyl substituted with from 1-3 Ra (e.g., trifluoromethyl).
10. The method of claim 9, wherein R1 is S(0)2CF3.
11. The method of any one of claims 1-5, wherein R4A is -(W4)q-W2.
12. The method of any one of claims 1-5 and 11, wherein q is 0.
13. The method of any one of claims 1-5 and 11, wherein q is 1.
14. The method of any one of claims 1-5, 11 and 13, wherein W1 is C1-3 alkylene (e.g.,
CH2).
15. The method of any one of claims 1-5 and 11-14, wherein W2 is C6-10 aryl, which is optionally substituted with from 1-4 Rc.
16. The method of any one of claims 1-5 and 11-15, wherein W2 is phenyl, which is optionally substituted with from 1-2 Rc (e.g., W2 is unsubstituted phenyl).
17. The method of any one of claims 1-5 and 11-14, wherein W2 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
18. The method of any one of claims 1-5, 11-14, and 17, wherein W2 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
19. The method of any one of claims 1-5, 11-14, and 17-18, wherein W2 is heteroaryl including from 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
20. The method of any one of claims 1-5, 11-14, and 17-19, wherein W2 is pyridinyl optionally substituted with from 1-2 independently selected Rc (e.g., W2 is pyridin-4-yl or pyridine-2 -yl, e.g., W2 is unsubstituted pyridin-4-yl or pyridin-2-yl).
21. The method of any one of claims 1-5, 11-14, and 17-18, wherein W2 is heteroaryl including from 5 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
22. The method of any one of claims 1-5, 11-14, 17-18, and 21, wherein W2 is thiazolyl optionally substituted with from 1-2 independently selected Rc (e.g., W2 is thiazol-5-yl optionally substituted with 1 Rc).
23. The method of any one of claims 1-5 and 11-22, wherein R1 is selected from:
Figure imgf000136_0001
24. The method of any one of claims 1-3, wherein R1 is NO2.
25. The method of any one of claims 1-2, wherein R1 is S(0)2N(R6A)2; or R1 is C(0)N(R6A)2; or wherein R1 is S(0)2NHR6A, such as S(0)2NH2 or S(0)2NH(CI-6 alkyl).
26. The method of any one of claims 1-25, wherein each of R6A is independently selected from:
(i) H; and (ii) Ci-6 alkyl optionally substituted with from 1-6 Ra (e.g., one R6A is H; and the other R6A is Ci-6 alkyl optionally substituted with from 1-6 Ra; e.g., R1 is S(0)2NHMe; or R1 is C(O)NHMe).
27. The method of any one of claims 1-2, wherein R1 is CN; or R1 is F.
28. The method of any one of claims 1-2, wherein R1 is C(=0)OR5A (e.g., wherein R5A can be selected from (i) H; and (ii) Ci-6 alkyl optionally substituted with from 1-6 Ra).
29. The method of claims 26 or 27, wherein R1 is C(=0)R4A (e.g., R4A can be H).
30. The method of any one of claims 1-29, wherein R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, NHC(0)R4B, OR5B, and SR5B; or wherein R2 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc, such as R2 is imidazolyl optionally substituted with one Rc
31. The method of any one of claims 1-30, wherein R2 is selected from the group consisting of: H, halo, cyano, OC(0)R4B, and SR5B (e.g., R2 can be selected from H, halo, and cyano).
32. The method of any one of claims 1-31, wherein each of R4B and R5B is independently selected from:
(ii) Ci-6 alkyl optionally substituted with 1-6 Ra; and
(iii) -(W4)q-W2 (e.g., q is 0; or q is 1).
33. The method of any one of claims 1-32, wherein each of R4B and R5B is independently selected from: (iii) -(W1)q-W2 (e.g., q is 0; or q is 1).
34. The method of claim 33, wherein q is 0.
35. The method of any one of claims 1-34, wherein W2 is selected from:
(b) C6-io aryl, which is optionally substituted with from 1-4 Rc;
(c) heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
36. The method of any one of claims 1-35, wherein W2 is phenyl, which is optionally
Figure imgf000138_0001
37. The method of any one of claims 1-35, wherein W2 is heteroaryl including from 5-
6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-4 independently selected Rc.
38. The method of any one of claims 1-35 and 37, wherein W2 is selected heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
39. The method of claim 38, wherein W2 is
Figure imgf000139_0001
(e.g., Rd is unsubstituted Ci-6 alkyl).
40. The method of any one of claims 33-36, wherein R4Bis phenyl, which is optionally
substituted with from
Figure imgf000139_0002
41. The method of any one of claims 33-34 and 38-39, wherein R5B is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with
from 1-2 independently selected
Figure imgf000139_0003
42. The method of claim 1, wherein R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
43. The method of any one of claims 1 and 42, wherein R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
44. The method of any one of claims 1 and 42-43, wherein R1 and R2 taken together with the carbon atoms to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring is substituted with from 1-2 oxo.
45. The method of any one of claims 1 and 42-44, wherein R1 and R2 taken together with the atoms to which each is attached forms a ring selected from the following:
Figure imgf000140_0001
46. The method of any one of claims 1-45, wherein each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl which is optionally substituted with from 1-2 Ra,
(iv) Ci-6 alkoxy which is optionally substituted with from 1-2 Ra, and
(v) Ci-6 haloalkoxy which is optionally substituted with from 1-2 Ra.
47. The method of any one of claims 1-46, wherein each of R3, R4, and R5 is independently selected from the group consisting of:
(i) H,
(ii) halo,
(iii) Ci-6 alkyl,
(iv) Ci-6 alkoxy, and
(v) Ci-6 haloalkoxy.
48. The method of any one of claims 1-47, wherein R3 is selected from the group consisting of: (i) H,
(ii) halo (e.g., F), and
(iii) Ci-6 alkyl (e.g., methyl).
49. The method of any one of claims 1-48, wherein R4 is selected from the group consisting of:
(i) H,
(ii) halo, and
(iv) Ci-6 alkoxy (e.g., methoxy).
50. The method of any one of claims 1-49, wherein R5 is H.
51. The method of any one of claims 1-45 and 50, wherein R3 and R4 taken together with the carbon to which each is attached forms a ring including from 3-8 ring atoms, wherein the ring includes: (a) from 2-8 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-3 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
52. The method of any one of claims 1-45 and 50-51, wherein R3 and R4 taken together with the carbon to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H, C1-3 alkyl, halo, hydroxy, and oxo; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
53. The method of any one of claims 1-45 and 50-52, wherein R3 and R4 taken together with the carbon to which each is attached forms a ring including from 5-6 ring atoms, wherein the ring includes: (a) from 2-6 ring carbon atoms, each of which is substituted with from 1-2 substituents independently selected from H; and (b) from 0-2 ring heteroatoms which are each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2.
54. The method of any one of claims 1-45 and 50-53, wherein R3 and R4 taken together with the carbon to which each is attached forms a ring selected from the following:
Figure imgf000142_0001
55. The method of any one of claims 1-54, wherein W is selected from:
(i) C(=0);
(ϋ) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo
(e g·, F).
56. The method of any one of claims 1-55, wherein W is C(=0).
57. The method of any one of claims 1-55, wherein W is C(=S).
58. The method of any one of claims 1-55, wherein W is C(=NRd) (e.g., Rd is Ci-6 alkyl, e.g., methyl).
59. The method of any one of claims 1-55, wherein W is C(=C-N02).
60. The method of any one of claims 1-55, wherein W is C1-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g., F) (e.g., unsubstituted C1-3 alkylene; or C1-3 alkylene substituted with from 1-4 fluoro).
61. The method of any one of claims 1-60, wherein Q and A are defined according to
(A).
62. The method of any one of claims 1-61, wherein Q is NH.
63. The method of any one of claims 1-62, wherein A is -(YA1)n-YA2.
64. The method of any one of claims 1-63, wherein n is 0.
65. The method of any one of claims 1-63, wherein n is 1.
66. The method of any one of claims 1-63 and 65, wherein YA1 is C1-3 alkylene (e.g., YA1 is CH2 or CH2CH2); or wherein YA1 is C1-3 alkylene substituted with oxo, such as CH2C(=0)-.
67. The method of any one of claims 1-66, wherein YA2 is C6-20 aryl, which is optionally substituted with from 1-4 Rc.
68. The method of any one of claims 1-67, wherein YA2 is C6-10 aryl, which is optionally substituted with from 1-3 Rc.
69. The method of any one of claims 1-68, wherein YA2 is phenyl, which is optionally substituted with from 1-3 Rc.
70. The method of any one of claims 1-69, wherein YA2 is phenyl which is substituted with 1 Rc.
71. The method of any one of claims 1-70, wherein YA2 is phenyl substituted with Rc at the para position.
72. The method of any one of claims 1-68, wherein YA2 is tetrahydro-naphthyl, which
is optionally substituted with from 1-3 Rc (e.g., YA2 can be
Figure imgf000144_0001
73. The method of any one of claims 1-66, wherein YA2 is heteroaryl including from 6- 10 (e.g., 8-10) ring atoms, wherein from 1-4 (e.g., from 1-2, e.g., 1) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc.
74. The method of claim 73, wherein YA2 heteroaryl including from 8-10 ring atoms, wherein from 1-2, such as 1, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently
Figure imgf000144_0002
wherein YA2 is heteroaryl including 5-6, such as 6 ring atoms, wherein from 1-2, such as 1, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heteroaryl ring carbon atoms are optionally substituted with from 1-2 independently selected Rc (e.g., YA2 is pyridyl which is optionally substituted with from 1-2 independently selected Rc).
75. The method of any one of claims 67-74, wherein each occurrence of Rc is independently selected from Ci-io alkyl which is optionally substituted with 1-2 independently selected Ra, Ci-4 alkoxy, and C3-6 cycloalkyl; or wherein each occurrence of Rc is independently C3-6 cycloalkyl such as cyclobutyl and cyclohexyl or C6-10 aryl such as phenyl.
76. The method of claim 75, wherein each Ra substituent of Rc is independently selected from hydroxy and Ci-6 alkoxy.
77. The method of any one of claims 75-76, wherein each occurrence of Rc is independently selected from butyl (e.g., n-butyl), propyl (e.g., n-propyl), cyclohexyl, 2- hydroxybut-l-yl, ethoxymethyl, methoxy, and ethoxy.
78. The method of any one of claims 1-77, wherein A is
Figure imgf000145_0001
(e.g., Rc is as defined in claims 75-77, e.g., Rc is n-butyl, cyclohexyl, 2- hydroxybut-l-yl, or ethoxymethyl).
79. The method of claim 1-77, wherein A is
Figure imgf000145_0002
defined in claims 75-77, e.g.,
Rc is ethoxy or propyl).
80. The method of any one of claims 1-77, wherein A is
Figure imgf000145_0003
Rc is as defined in claims 75-77, e.g., Rc is ethoxy).
81. The method of any one of claims 1-66, wherein YA2 is C3-20 cycloalkyl, which is optionally substituted with from 1-4 Rb.
82. The method of any one of claims 1-66 and 81, wherein YA2 is C5-10 cycloalkyl, which is optionally substituted with from 1-4 Rb.
83. The method of any one of claims 1-66 and 81-82, wherein YA2 is C6-8 cycloalkyl such as cyclohexyl, which is optionally substituted with from 1-2 Rb.
84. The method of any one of claims 81-83, wherein each occurrence of Rb is selected from Ci-io alkyl optionally substituted with from 1-6 independently selected Ra; Ci-4 haloalkyl; -OH; oxo; -F; -Cl; -Br; Ci-4 alkoxy; Ci-4 haloalkoxy; and C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl; or wherein each occurrence of Rb is C3-6 cycloalkyl optionally substituted with from 1-4 independently selected Ci-4 alkyl or C6-10 aryl.
85. The method of any one of claims 81-84, wherein each occurrence of Rb is Ci-10 alkyl optionally substituted with from 1-6 independently selected Ra; Ci-4 haloalkyl; -F; - Cl; -Br; and Ci-4 alkoxy.
86. The method of any one of claims 81-85, wherein each occurrence of Rb is Ci-6 alkyl optionally substituted with from 1-3 independently selected Ra; -F; and Ci-4 alkoxy.
87. The method of any one of claims 81-86, wherein each occurrence of Rb is n-propyl; n-butyl; ethoxymethyl; -F; and Ci-4 alkoxy (e.g., -F, n-propyl, and ethoxymethyl).
88 The method of any one of claims 81-87, wherein A is selected from:
Figure imgf000146_0001
89. The method of claim 88, wherein Rb is as defined in claims 84-87 (e.g., Rb is F; or
Rb is n-propyl; or Rb is ethoxymethyl).
90. The method of any one of claims 1-61, wherein Q is N(Rq).
91. The method of any one of claims 1-61 and 90, wherein Rq and R4, taken together with the atoms connecting them, forms a ring including 5-8 ring atoms, wherein the ring includes (a) from 2-7 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each heteroatom is independently selected from N, N(H), O, and S(0)o-2.
92. The method of any one of claims 1-61 and 91, wherein Rq and R4, taken together with the atoms connecting them, forms a ring including 5-6 ring atoms, wherein the ring includes (a) from 2-5 carbon atoms and (b) from 0-2 heteroatoms aside from Q, wherein each heteroatom is independently selected from N, N(H), O, and S(0)o-2.
93. The method of any one of claims 1-60, wherein Q and A are defined according to
(B).
94. The method of any one of claims 1-60 and 93, wherein E is heterocyclyl including from 3-12 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1-2 independently selected Rb.
95. The method of any one of claims 1-60 and 93-94, wherein E is heterocyclyl including from 3-12 ring atoms, wherein aside from the nitrogen atom present, from 0-3 additional ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein one or more of the heterocyclyl ring carbon atoms are optionally substituted with from 1 independently selected Rb.
96. The method of any one of claims 1-60 and 93-95, wherein E is:
Figure imgf000148_0001
97. The method of claim 1, wherein the compound is of Formula (1-1):
Figure imgf000148_0002
wherein Rcl is as defined for Rc.
98. The method of claim 97, wherein Rcl is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
99. The method of claim 1, wherein the compound is of Formula (1-2):
Figure imgf000148_0003
wherein Rc2 is as defined for Rc.
100. The method of claim 99, wherein Rc2 is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
101. The method of claim 1, wherein the compound is of Formula (I-3a), (I-3b), or (I- 3c):
Figure imgf000149_0001
wherein each Rb3 is an independently selected Rb.
102. The method of claim 101, wherein each Rb3 is an independently selected Rb; and wherein said Rb is as defined in any one of claims 84-87.
103. The method of claim 1, wherein the compound is of Formula (1-4)
Figure imgf000149_0002
wherein Rc4 is as defined for Rc.
104. The method of claim 103, wherein Rc4 is as defined for Rc; and wherein said Rc is as defined in any one of claims 75-77.
105. The method of claim 1, wherein the compound is of Formula (1-5)
Figure imgf000150_0001
wherein Rc5 is as defined for Rc.
106. The method of claim 1, wherein the compound is of Formula (1-6):
Figure imgf000150_0002
107. The method of claim 97-106, wherein W is selected from:
(i) C(=0);
(ii) C(=S);
(iii) C(=NRd);
(v) S(0)2;
(vii) S(0)(NH);
(viii) C(=C-N02); and
(ix) Ci-3 alkylene optionally substituted with from 1-4 independently selected halo (e.g., F).
108. The method of any one of claims 97-107, wherein W is C(=0).
109. The method of any one of claims 97-108, wherein R1 is as defined in any one of claims 2-29 (e.g., R1 is N02).
110. The method of any one of claims 97-109, wherein R2 is as defined in any one of claims 30-41.
111. The method of any one of claims 97-110, wherein R1 and R2 are as defined in any one of claims 42-45.
112. The method of any one of claims 97-111, wherein each of R3, R4, and R5 is as defined in any one of claims 46-47 (e.g., R5 is H).
113. The method of any one of claims 97-112, wherein R3 is as defined in claim 48.
114. The method of any one of claims 97-113, wherein R4 is as defined in claim 49.
115. The method of any one of claims 97-114, wherein R3 and R4 is as defined in any one of claims 51-54.
116. The method of claim 1, wherein the compound is selected from the following:
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
or a pharmaceutically acceptable salt thereof.
117. The method of any one of claims 46-96, wherein R1 is S02R4A or S(0)I-2N(R6A)2; and R2 is halo such as chloro.
118. The method of any one of claims 46-96, wherein R1 is CN or C(=0)R4A; and R2 is halo such as chloro.
119. The method of any one of claims 1-118, wherein the inhibiting comprises antagonizing STING.
120. The method of any one of claims 1-119, which is carried out in vitro.
121. The method of claim 120, wherein the method comprises contacting a sample comprising one or more cells comprising STING with the compound.
122. The method of claim 121, wherein the one or more cells are one or more cancer cells.
123. The method of claim 121 or 122 wherein the sample further comprises one or more cancer cells, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.
124. The method of any one of claims 1-118, which is carried out in vivo.
125. The method of claim 124, wherein the method comprises administering the compound to a subject having a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease.
126. The method of claim 125, wherein the subject is a human.
127. The method of claim 125, wherein the disease is cancer.
128. The method of claim 127, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.
129. The method of claim 127 or 128, wherein the cancer is a refractory cancer.
130. The method of claim 125, wherein the compound is administered in combination with one or more additional cancer therapies.
131. The method of claim 130, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.
132. The method of claim 131, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.
133. The method of claim 132, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g.,azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisom erase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an anti helminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD-
1 - PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL-10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein
(LAG3), MHC class II - LAG3, 4-1BB-4-1BB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM
- LIGHT, HVEM-BTL A-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48
- CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2,
HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PDl or PD-L1).
134. The method of any one of claims 125-133, wherein the compound is administered intratumorally.
135. A method of treating cancer, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula (I) as claimed in claim 1
136. The method of claim 135, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.
137. The method of claim 135 or 136, wherein the cancer is a refractory cancer.
138. The method of claim 135, wherein the compound is administered in combination with one or more additional cancer therapies.
139. The method of claim 138, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.
140. The method of claim 139, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.
141. The method of claim 140, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g.,azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisom erase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracy clines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an anti- helminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD-
1 - PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL-10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT -LT A, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM
- LIGHT, HVEM-BTL A-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48
- CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD 160, CD30, and CD 155 (e.g., CTLA-4 or PD1 or PD-L1).
142. The method of any one of claims 135-141, wherein the compound is administered intratumorally.
143. A method of inducing an immune response in a subj ect in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) as claimed in claim 1 .
144. The method of claim 143, wherein the subject has cancer.
145. The method of claim 144, wherein the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies.
146. The method of claim 144, wherein the cancer selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma .
147. The method of claim 146, wherein the cancer is a refractory cancer.
148. The method of claim 143, wherein the immune response is an innate immune response.
149. The method of claim 145, wherein the at least one or more cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.
150. The method of claim 149, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.
151. The method of claim 150, wherein the one or more additional chemotherapeutic agents is selected from alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g.,azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisom erase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an anti helminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD- 1 - PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL-10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein
(LAG3), MHC class II - LAG3, 4-1BB-4-1BB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM
- LIGHT, HVEM-BTL A-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48
- CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2,
HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PDl or PD-L1).
152. A method of treatment of a disease in which increased (e.g., excessive) STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the disease, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula (I) as claimed in claim 1 .
153. A method of treatment comprising administering to a subject having a disease in which increased (e.g., excessive) STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the disease an effective amount of a compound of Formula (I) as claimed in claim 1 .
154. A method of treatment comprising administering to a subject a compound of Formula (I) as claimed in claim 1 , wherein the compound or composition is administered in an amount effective to treat a disease in which increased (e.g., excessive) STING or cGAS signaling contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.
155. The method of any one of claims 152-154, wherein the disease is cancer.
156. The method of claim 155, wherein the cancer is selected from the group consisting of melanoma, cervical cancer, breast cancer, ovarian cancer, prostate cancer, testicular cancer, urothelial carcinoma, bladder cancer, non-small cell lung cancer, small cell lung cancer, sarcoma, colorectal adenocarcinoma, gastrointestinal stromal tumors, gastroesophageal carcinoma, colorectal cancer, pancreatic cancer, kidney cancer, hepatocellular cancer, malignant mesothelioma, leukemia, lymphoma, myelodysplasia syndrome, multiple myeloma, transitional cell carcinoma, neuroblastoma, plasma cell neoplasms, Wilm's tumor, or hepatocellular carcinoma.
157. The method of claim 155 or 156, wherein the cancer is a refractory cancer.
158. The method of any one of claims 155-157, wherein the compound is administered in combination with one or more additional cancer therapies.
159. The method of claim 158, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.
160. The method of claim 159, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.
161. The method of claim 160, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g.,azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisom erase; e.g., camptothecins, such as irinotecan and/or topotecan;. amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti- angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an anti- helminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1 - PD-L1, PD-
1 - PD-L2, interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL-10, transforming growth factor-b (TGFP), T cell immunoglobulin and mucin 3 (TIM3 or HAVCR2), Galectin 9 - TIM3, Phosphatidylserine - TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II - LAG3, 4- 1BB-4- IBB ligand, 0X40-0X40 ligand, GITR, GITR ligand - GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40- CD40 ligand, HVEM-LIGHT -LT A, HVEM, HVEM - BTLA, HVEM - CD 160, HVEM
- LIGHT, HVEM-BTL A-CD 160, CD80, CD80 - PDL-1, PDL2 - CD80, CD244, CD48
- CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, KIRs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86 - CD28, CD86 - CTLA, CD80 - CD28, CD39, CD73 Adenosine-CD39- CD73, CXCR4-CXCL12, Phosphatidylserine, TIM3, Phosphatidylserine - TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PDl or PD-L1).
162. The method of any one of claims 152-161, wherein the compound is administered intratumorally.
163. A method of treatment of a disease, disorder, or condition associated with STING or cGAS, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula (I) as claimed in claim 1 .
164. The method of claim 163, wherein the disease, disorder, or condition is selected from type I interferonopathies, Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, inflammation-associated disorders, and rheumatoid arthritis.
165. The method of claim 164, wherein the disease, disorder, or condition is a type I interferonopathy (e.g., STING-associated vasculopathywith onset in infancy (SAVI)).
166. The method of claim 165, wherein the type I interferonopathy is STING- associated vasculopathy with onset in infancy (SAVI)).
167. The method of claim 164, wherein the disease, disorder, or condition is Aicardi- Goutieres Syndrome (AGS).
168. The method of claim 164, wherein the disease, disorder, or condition is a genetic form of lupus.
169. The method of claim 164, wherein the disease, disorder, or condition is inflammation-associated disorder.
170. The method of claim 169, wherein the inflammation-associated disorder is systemic lupus erythematosus.
171. The method of any one of claims 118-170, wherein the method further comprises identifying the subject.
172. The method of any one of the preceding claims, wherein the method further comprises administering the compound as a pharmaceutical composition which comprises a compound of Formula (I) as claimed in any one or claims 1-118 and one or more pharmaceutically acceptable excipients.
173. A pharmaceutical composition which comprises a compound of Formula (I) as claimed in claim 1 and a pharmaceutically acceptable excipient.
174. The method of any one of claims 119-172, wherein the method comprises administering a compound of Formula (I) as claimed in any one or claims 2-118.
PCT/US2019/062245 2018-11-19 2019-11-19 Compounds and compositions for treating conditions associated with sting activity WO2020106741A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862769500P 2018-11-19 2018-11-19
US62/769,500 2018-11-19
US201962861108P 2019-06-13 2019-06-13
US62/861,108 2019-06-13

Publications (1)

Publication Number Publication Date
WO2020106741A1 true WO2020106741A1 (en) 2020-05-28

Family

ID=68919767

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/062245 WO2020106741A1 (en) 2018-11-19 2019-11-19 Compounds and compositions for treating conditions associated with sting activity

Country Status (1)

Country Link
WO (1) WO2020106741A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020257621A1 (en) 2019-06-21 2020-12-24 Ifm Due, Inc. Methods of treating cancer
WO2022140410A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2022140403A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2022140397A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2022140387A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
CN115806502A (en) * 2022-12-29 2023-03-17 杭州百凯生物医药有限公司 Alpha-amidone derivative and preparation method and application thereof
US11618749B2 (en) 2018-07-03 2023-04-04 Ifm Due, Inc. Compounds and compositions for treating conditions associated with STING activity
WO2023164201A1 (en) 2022-02-28 2023-08-31 Astellas Pharma, Inc. Aryl alkynamide derivatives
WO2024064358A1 (en) 2022-09-23 2024-03-28 Ifm Due, Inc. Compounds and compositions for treating conditions associated with sting activity

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002051397A1 (en) * 2000-12-22 2002-07-04 Ishihara Sangyo Kaisha, Ltd. Aniline derivatives or salts thereof and cytokine production inhibitors containing the same
WO2005032493A2 (en) * 2003-10-07 2005-04-14 Renovis, Inc. Amide compounds as ion channel ligands and uses thereof
US7927613B2 (en) 2002-02-15 2011-04-19 University Of South Florida Pharmaceutical co-crystal compositions
US20120202848A1 (en) 2010-12-03 2012-08-09 The Trustees Of The University Of Pennsylvania Therapy of autoimmune colitis using a tip60 inhibitor
WO2013093095A1 (en) * 2011-12-22 2013-06-27 Kancera Ab Bisarylsulfonamides useful in the treatment of inflammation and cancer
WO2015061294A2 (en) 2013-10-21 2015-04-30 Philadelphia Health & Education Corporation D/B/A/ Use of sting agonists to treat chronic hepatitis b virus infection
CN106496108A (en) * 2016-11-01 2017-03-15 上海应用技术大学 There is amides compound and its application of anti-tumor activity
WO2017123657A1 (en) * 2016-01-11 2017-07-20 Gary Glick Cyclic dinucleotides for treating conditions associated with sting activity such as cancer
WO2017175156A1 (en) * 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002051397A1 (en) * 2000-12-22 2002-07-04 Ishihara Sangyo Kaisha, Ltd. Aniline derivatives or salts thereof and cytokine production inhibitors containing the same
US7927613B2 (en) 2002-02-15 2011-04-19 University Of South Florida Pharmaceutical co-crystal compositions
WO2005032493A2 (en) * 2003-10-07 2005-04-14 Renovis, Inc. Amide compounds as ion channel ligands and uses thereof
US20120202848A1 (en) 2010-12-03 2012-08-09 The Trustees Of The University Of Pennsylvania Therapy of autoimmune colitis using a tip60 inhibitor
WO2013093095A1 (en) * 2011-12-22 2013-06-27 Kancera Ab Bisarylsulfonamides useful in the treatment of inflammation and cancer
WO2015061294A2 (en) 2013-10-21 2015-04-30 Philadelphia Health & Education Corporation D/B/A/ Use of sting agonists to treat chronic hepatitis b virus infection
WO2017123657A1 (en) * 2016-01-11 2017-07-20 Gary Glick Cyclic dinucleotides for treating conditions associated with sting activity such as cancer
WO2017175156A1 (en) * 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators
CN106496108A (en) * 2016-11-01 2017-03-15 上海应用技术大学 There is amides compound and its application of anti-tumor activity

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
"Handbook of Pharmaceutical Additives", 2007, GOWER PUBLISHING COMPANY
"Pharmaceutical Preformulation and Formulation", 2009, THE PHARMACEUTICAL PRESS
"Remington: The Science and Practice of Pharmacy", 2012, LIPPINCOTT WILLIAMS & WILKINS
FILIPSKI, K.J. ET AL., CURRENT TOPICS IN MEDICINAL CHEMISTRY, vol. 13, 2013, pages 776 - 802
HAAG SIMONE M ET AL: "Targeting STING with covalent small-molecule inhibitors", NATURE, MACMILLAN JOURNALS LTD, LONDON, vol. 559, no. 7713, 4 July 2018 (2018-07-04), pages 269 - 273, XP036553086, ISSN: 0028-0836, [retrieved on 20180704], DOI: 10.1038/S41586-018-0287-8 *
L. FIESERM. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
LAMMERS ET AL.: "Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems", NEOPLASIA, vol. 10, 2006, pages 788 - 795
LIU WANG-QING ET AL: "NRPa-308, a new neuropilin-1 antagonist, exertsin vitroanti-angiogenic and anti-proliferative effects andin vivoanti-cancer effects in a mouse xenograft model", CANCER LETTERS, vol. 414, 21 February 2018 (2018-02-21), pages 88 - 98, XP085319977, ISSN: 0304-3835, DOI: 10.1016/J.CANLET.2017.10.039 *
POSTOW, M. J., CLIN. ONCOL., vol. 33, 2015, pages 1
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
T. W. GREENERGM. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11618749B2 (en) 2018-07-03 2023-04-04 Ifm Due, Inc. Compounds and compositions for treating conditions associated with STING activity
WO2020257621A1 (en) 2019-06-21 2020-12-24 Ifm Due, Inc. Methods of treating cancer
WO2022140410A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2022140403A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2022140397A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2022140387A1 (en) 2020-12-22 2022-06-30 Ifm Due, Inc. Methods of treating cancer
WO2023164201A1 (en) 2022-02-28 2023-08-31 Astellas Pharma, Inc. Aryl alkynamide derivatives
US11912679B2 (en) 2022-02-28 2024-02-27 Astellas Pharma, Inc. Aryl alkynamide derivatives
WO2024064358A1 (en) 2022-09-23 2024-03-28 Ifm Due, Inc. Compounds and compositions for treating conditions associated with sting activity
CN115806502A (en) * 2022-12-29 2023-03-17 杭州百凯生物医药有限公司 Alpha-amidone derivative and preparation method and application thereof
CN115806502B (en) * 2022-12-29 2024-02-23 杭州百凯生物医药有限公司 Alpha-amidoketone derivative and preparation method and application thereof

Similar Documents

Publication Publication Date Title
US11618749B2 (en) Compounds and compositions for treating conditions associated with STING activity
WO2020150417A2 (en) Compounds and compositions for treating conditions associated with sting activity
WO2020010155A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2020106741A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2020252240A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2020150439A1 (en) Compounds and compositions for treating conditions associated with sting activity
EP3976584A1 (en) Compounds and compositions for treating conditions associated with sting activity
EP3883651A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2020236586A1 (en) N-hetaryl-squaramide compounds for treating conditions associated with sting activity
WO2021067801A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2021067791A1 (en) Oxalamide compounds and compositions for treating conditions associated with sting activity
WO2021067805A1 (en) Oxalamide heterobycyclic compounds and compositions for treating conditions associated with sting activity
EP4182030A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2022150585A1 (en) Heterobicyclic compounds having an urea or analogue and their compositions for treating conditions associated with sting activity
US20220024919A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2022015938A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2023137034A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2022150543A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2022150549A1 (en) Oxalamide compounds and compositions for treating conditions associated with sting activity
WO2022150560A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2022133098A2 (en) Compounds and compositions for treating conditions associated with sting activity
US20240076285A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2023137041A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2024064358A1 (en) Compounds and compositions for treating conditions associated with sting activity

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19821458

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19821458

Country of ref document: EP

Kind code of ref document: A1