COMPOUNDS AND COMPOSITIONS FOR TREATING CONDITIONS ASSOCIATED WITH STING ACTIVITY

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 (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING 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
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 (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING 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

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 TRF-3 to induce type I interferons (IFNs) and other co-regulated antiviral factors. In addition, STING was shown to trigger NF-κB 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 (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING 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.


An “antagonist” of STING 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.


In one aspect, compounds of Formula (I), or a pharmaceutically acceptable salt thereof, are featured:




embedded image


in which Q1, LA, Y1, Y2, Y3, X1, X2, R6, and W can be as defined anywhere herein.


In one aspect, pharmaceutical compositions are featured that include a chemical entity described herein (e.g., a compound described generically 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 (e.g., antagonizing) STING activity are featured that include contacting STING with a chemical entity described herein (e.g., a 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 (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 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 are featured, e.g., treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING 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 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 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-associated conditions are featured, e.g., type I interferonopathies (e.g., STING-associated vasculopathy with 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 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 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 activation (e.g., STING 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 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 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 activation (e.g., STING 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 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 activation (e.g., STING signaling) contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.


In another aspect, there is provided is a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein, for use in the treatment of a disease, condition or disorder modulated by STING inhibition.


In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of a condition, disease or disorder associated with increased (e.g., excessive) STING activation.


In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, described herein for use in the treatment of cancer.


In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of 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.


In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of type I interferonopathies.


In another aspect, there is provided a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the treatment of type I interferonopathies selected from STING-associated vasculopathy with 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 another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of a condition, disease or disorder associated with increased (e.g., excessive) STING activation.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of cancer.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of 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.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein in the manufacture of a medicament for the treatment of type I interferonopathies.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for use in the manufacture of a medicament for the treatment of type I interferonopathies selected from STING-associated vasculopathy with 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 another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein, for the treatment of a disease, condition or disorder modulated by STING inhibition.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of a condition, disease or disorder associated with increased (e.g., excessive) STING activation.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of cancer.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of 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.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of type I interferonopathies.


In another aspect, there is provided the use of a compound, or a pharmaceutically acceptable salt or tautomer thereof, as described herein for the treatment of type I interferonopathies selected from STING-associated vasculopathy with onset in infancy (SAVI)), Aicardi-Goutieres Syndrome (AGS), genetic forms of lupus, and inflammation-associated disorders such as systemic lupus erythematosus, and rheumatoid arthritis.


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-associated conditions, e.g., type I interferonopathies (e.g., STING-associated vasculopathy with 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 topoisomerase; 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, Muromonab-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-β (TGFβ), 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, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD160, HVEM-LIGHT, HVEM-BTLA-CD160, 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 PD1 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.


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 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 et 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, Fla., 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-methyl-D-glucamine, 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 saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C1-10 indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, iso-propyl, tert-butyl, n-hexyl. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.


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., —OCH3).


The term “alkylene” refers to a divalent alkyl (e.g., —CH2—).


The term “alkenyl” refers to an acyclic 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. Alkenyl groups can either be unsubstituted or substituted with one or more substituents.


The term “alkynyl” refers to an acyclic 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. Alkynyl groups can either be unsubstituted or substituted with one or more substituents.


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, dihydro-1H-indenyl and the like.


The term “cycloalkyl” as used herein refers to cyclic saturated hydrocarbon groups having, e.g., 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]butanyl, bicyclo[2.1.0]pentanyl, bicyclo[1.1.1]pentanyl, bicyclo[3.1.0]hexanyl, bicyclo[2.1.1]hexanyl, bicyclo[3.2.0]heptanyl, bicyclo[4.1.0]heptanyl, bicyclo[2.2.1]heptanyl, bicyclo[3.1.1]heptanyl, bicyclo[4.2.0]octanyl, bicyclo[3.2.1]octanyl, bicyclo[2.2.2]octanyl, 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]pentanyl, spiro[2.5]octanyl, spiro[3.5]nonanyl, spiro[3.5]nonanyl, spiro[3.5]nonanyl, spiro[4.4]nonanyl, spiro[2.6]nonanyl, spiro[4.5]decanyl, spiro[3.6]decanyl, spiro[5.5]undecanyl, and the like. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms.


The term “cycloalkenyl” as used herein means 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. As partially unsaturated cyclic hydrocarbon groups, cycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, 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, and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl). 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-d]pyrimidinyl, pyrrolo[2,3-b]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-b]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridinyl, pyrazolo[4,3-b]pyridinyl, tetrazolyl, chromanyl, 2,3-dihydrobenzo[b][1,4]dioxinyl, benzo[d][1,3]dioxolyl, 2,3-dihydrobenzofuranyl, tetrahydroquinolinyl, 2,3-dihydrobenzo[b][1,4]oxathiinyl, isoindolinyl, 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 saturated 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]butanyl, 2-azabicyclo[2.1.0]pentanyl, 2-azabicyclo[1.1.1]pentanyl, 3-azabicyclo[3.1.0]hexanyl, 5-azabicyclo[2.1.1]hexanyl, 3-azabicyclo[3.2.0]heptanyl, octahydrocyclopenta[c]pyrrolyl, 3-azabicyclo[4.1.0]heptanyl, 7-azabicyclo[2.2.1]heptanyl, 6-azabicyclo[3.1.1]heptanyl, 7-azabicyclo[4.2.0]octanyl, 2-azabicyclo[2.2.2]octanyl, 3-azabicyclo[3.2.1]octanyl, 2-oxabicyclo[1.1.0]butanyl, 2-oxabicyclo[2.1.0]pentanyl, 2-oxabicyclo[1.1.1]pentanyl, 3-oxabicyclo[3.1.0]hexanyl, 5-oxabicyclo[2.1.1]hexanyl, 3-oxabicyclo[3.2.0]heptanyl, 3-oxabicyclo[4.1.0]heptanyl, 7-oxabicyclo[2.2.1]heptanyl, 6-oxabicyclo[3.1.1]heptanyl, 7-oxabicyclo[4.2.0]octanyl, 2-oxabicyclo[2.2.2]octanyl, 3-oxabicyclo[3.2.1]octanyl, 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]pentanyl, 4-azaspiro[2.5]octanyl, 1-azaspiro[3.5]nonanyl, 2-azaspiro[3.5]nonanyl, 7-azaspiro[3.5]nonanyl, 2-azaspiro[4.4]nonanyl, 6-azaspiro[2.6]nonanyl, 1,7-diazaspiro[4.5]decanyl, 7-azaspiro[4.5]decanyl 2,5-diazaspiro[3.6]decanyl, 3-azaspiro[5.5]undecanyl, 2-oxaspiro[2.2]pentanyl, 4-oxaspiro[2.5]octanyl, 1-oxaspiro[3.5]nonanyl, 2-oxaspiro[3.5]nonanyl, 7-oxaspiro[3.5]nonanyl, 2-oxaspiro[4.4]nonanyl, 6-oxaspiro[2.6]nonane, 1,7-dioxaspiro[4.5]decanyl, 2,5-dioxaspiro[3.6]decanyl, 1-oxaspiro[5.5]undecanyl, 3-oxaspiro[5.5]undecanyl, 3-oxa-9-azaspiro[5.5]undecanyl and the like. The term “saturated” as used in this context means only single bonds present between constituent ring atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.


The term “heterocycloalkenyl” as used herein means partially unsaturated cyclic 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 heterocycloalkenyl groups include, without limitation, tetrahydropyridyl, dihydropyrazinyl, dihydropyridyl, dihydropyrrolyl, dihydrofuranyl, dihydrothiophenyl. As partially unsaturated cyclic groups, heterocycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the heterocycloalkenyl group is not fully saturated overall. Heterocycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.


As used herein, when a ring is described as being “aromatic”, it means said ring has a continuous, delocalized π-electron system. Typically, the number of out of plane 71-electrons corresponds to the Hückel rule (4n+2). Examples of such rings include: benzene, pyridine, pyrimidine, pyrazine, pyridazine, pyridone, pyrrole, pyrazole, oxazole, thioazole, isoxazole, isothiazole, and the like.


As used herein, when a ring is described as being “partially unsaturated”, it means said ring has one or more additional degrees of unsaturation (in addition to the degree of unsaturation attributed to the ring itself, e.g., one or more double or tirple bonds between constituent ring atoms), provided that the ring is not aromatic. Examples of such rings include: cyclopentene, cyclohexene, cycloheptene, dihydropyridine, tetrahydropyridine, dihydropyrrole, dihydrofuran, dihydrothiophene, and the like.


For the avoidance of doubt, and unless otherwise specified, for rings and cyclic groups (e.g., aryl, heteroaryl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, cycloalkyl, and the like described herein) containing a sufficient number of ring atoms to form bicyclic or higher order ring systems (e.g., tricyclic, polycyclic ring systems), it is understood that such rings and cyclic groups encompass those having fused rings, including those in which the points of fusion are located (i) on adjacent ring atoms (e.g., [x.x.0] ring systems, in which 0 represents a zero atom bridge (e.g.,




embedded image


(ii) a single ring atom (spiro-fused ring systems) (e.g.,




embedded image


or (iii) a contiguous array of ring atoms (bridged ring systems having all bridge lengths>0) (e.g.,




embedded image


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 moiety:




embedded image


encompasses the tautomeric form containing the moiety:




embedded image


Similarly, a pyridinyl or pyrimidinyl moiety that is described to be optionally substituted with hydroxyl encompasses pyridone or pyrimidone tautomeric forms.


As used herein, the phrase “optionally substituted” when used in conjunction with a structural moiety (e.g., alkyl) is intended to encompass both the unsubstituted structural moiety (i.e., none of the substitutable hydrogen atoms are replaced with one or more non-hydrogen substituents) and substituted structural moieties substituted with the indicated range of non-hydrogen substituents. For example, “C1-C4 alkyl optionally substituted with 1-4 Ra” is intended to encompass both unsubstituted C1-C4 alkyl and C1-C4 alkyl substituted with 1-4 Ra.


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 (e.g., antagonize) Stimulator of Interferon Genes (STING). Said chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) STING activation (e.g., STING 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 I Compounds


In one aspect, the disclosure features A compound of Formula (I):




embedded image


or a pharmaceutically acceptable salt thereof or a tautomer thereof, wherein:


LA is -(L1)a1-(L2)a2-(L3)a3-(L4)a4-(L5)a5-*, wherein * represents the point of attachment to Q1;


a1, a2, a3, a4, and a5 are each independently 0 or 1,


provided that a1+a2+a3+a4+a5≥1, and


each of L1, L3, and L5 is independently selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, S(O)0-2, and —C(═O)—;


provided that when one or both of a2 and a4 is 0, then the combinations of L1, L3, and L5 cannot form O—O, N—O, N—N, O—S, S—S, or N—S(O)0 bonds, and


further provided that LA cannot include a cyclic group directly attached to the 6-membered ring containing Y1, Y2, and Y3;


each of L2 and L4 is independently selected from the group consisting of:

    • straight-chain C1-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb.
    • C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc provided the C3-10 cycloalkylene or C3-10 cycloalkenylene is not directly connected to the 6-membered ring containing Y1, Y2, and Y3; and
    • heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc, provided the heterocyclylene or heterocycloalkenylene is not directly connected to the 6-membered ring containing Y1, Y2, and Y3;
    • Q1 is —Rg;


Y1, Y2, and Y3 are each independently selected from the group consisting of CR1, C(═O), N, and NR2;


X1 is selected from the group consisting of O, S, N, NR2, and CR1;


X2 is selected from the group consisting of O, S, N, NR4, and CR5;


each custom-character is independently a single bond or a double bond, provided that the five-membered ring comprising X1 and X2 is heteroaryl, and that the six-membered ring comprising Y1, Y2, and Y3 is aryl or heteroaryl;


each occurrence of R1 and R5 is independently selected from the group consisting of: H; Rc; Rg; and -(Lg)bg-Rg;


each occurrence of R2 and R4 is independently selected from the group consisting of: H; Rd; Rg; and -(Lg)bg-Rg;


R6 is selected from the group consisting of: H; Rd; and Rg;


W is selected from the group consisting of:




embedded image


Ring B1 is a heteroarylene of 5 ring atoms, wherein 1-4 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S; wherein the heteroarylene of Ring B1 is optionally substituted with 1-2 substituents independently selected from the group consisting of oxo and Rc, provided that Ring B1 is attached to the C(═O)NR6 group via a ring carbon atom;


each LAA is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-2 Ra; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa1 is 0, 1, or 2;


Ring C1 is selected from the group consisting of:

    • C3-12 cycloalkylene or C3-12 cycloalkenylene, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and (LAA)aa1-Rg;
    • heterocyclylene or heterocycloalkenylene of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and (LAA)aa1-Rg;
    • heteroarylene of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroarylene is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc and (LAA)aa1-Rg; and
    • C6-10 arylene optionally substituted with 1-4 substituents independently selected from the group consisting of Rc and (LAA)aa1-Rg;


R7 is selected from the group consisting of: Rg and -(L7)b7-Rg;


each L7 is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-2 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O); and


b7 is 1, 2, or 3;




embedded image


Ring B2 is a heteroarylene of 5 ring atoms, wherein 1-4 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S, wherein the heteroarylene of Ring B is optionally substituted with 1-2 substituents independently selected from the group consisting of: oxo and Rc, provided that Ring B is attached to the C(═O)NR6 group via a ring carbon atom;


each LAB is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa2 is 0, 1, 2, or 3;


Ring C2 is selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;
    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; and
    • C6-10 aryl optionally substituted with 1-4 Rc;


(iii) heteroaryl of 5 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; provided the heteroaryl is attached to the C(═O)NR6 group via a ring carbon atom;




embedded image


P1, P2, P3, P4, and P5 are each independently selected from the group consisting of: N, NH, NRd, NR71, CH, CRc, CR71, and C(═O), provided that 1-3, such as 1, of P2, P3, and P4 is CR71 or NR71;


each occurrence of R71 is independently -(LAC)aa3-R8, wherein:


each LAC is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra; —O—; —NRN; —S(O)0-2; C(O); C(O)O; OC(O); NRNC(O); C(O)NRN; NRNC(O)NRN; NRNC(O)O; and OC(O)NRN;


aa3 is 0, 1, 2, or 3;


each occurrence of R8 is independently Rg or C1-10 alkyl optionally substituted with 1-6 Ra1; and


each occurrence of RN is independently H or Rd;


(v) a bicyclic or polycyclic ring system selected from the group consisting of:

    • bicyclic or polycyclic C5-15 cycloalkyl or C5-15 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heterocyclyl or heterocycloalkenyl of 7-15 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heteroaryl of 8-15 ring atoms, wherein 1-6 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of: oxo, Rc, and -(LAD)bB-Rg; and
    • bicyclic or polycyclic C8-15 aryl optionally substituted with 1-4 substituents independently selected from the group consisting of: oxo, Rc, and -(LAD)bB-Rg,
    • provided the bicyclic or polycyclic heteroring is attached to the C(═O)NR6 group via a ring carbon atom;
    • each occurrence of LAD is selected from the group consisting of: —O—, —NH—, —NRd, —S(O)0-2, C(O), and C1-3 alkylene optionally substituted with 1-3 Ra; and
      • bB is 0, 1, 2, or 3;
      • AND




embedded image


LAE is selected from the group consisting of:

    • C1-6 alkylene, C2-6 alkenylene, or C2-6 alkynylene, each of which is optionally substituted with 1-6 Ra;
    • monocyclic C3-8 cycloalkylene or C3-8 cycloalkenylene, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; and
    • monocyclic heterocyclylene or heterocycloalkenylene of 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc, provided that the heterocycloylene or heterocycloalkenylene is attached to the C(═O)NR6 group via a ring carbon atom;


each LAF is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa4 is 0, 1, 2, or 3; and


Ring C4 is Rg;


each occurrence of Ra and is independently selected from the group consisting of: —OH; -halo; —NReRf; C1-4 alkoxy; C1-4 haloalkoxy; —C(═O)O(C1-4 alkyl); —C(═O)(C1-4 alkyl); —C(═O)OH; —CONR′R″; —S(O)1-2NR′R″; —S(O)1-2(C1-4 alkyl); and cyano;


each occurrence of Rb and Rc is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy; C1-4 haloalkoxy; —S(O)1-2(C1-4 alkyl); —S(O)(═NH)(C1-4 alkyl); —NReRf; —OH; —S(O)1-2NR′R″; —C1-4 thioalkoxy; —NO2; —C(═O)(C1-10 alkyl); —C(═O)O(C1-4 alkyl); —C(═O)OH; —C(═O)NR′R″; and —SF5;


each occurrence of Rd is independently selected from the group consisting of: C1-6 alkyl optionally substituted with 1-3 independently selected Ra; —C(O)(C1-4 alkyl); —C(O)O(C1-4 alkyl); —CONR′R″; —S(O)1-2NR′R″; —S(O)1-2(C1-4 alkyl); —OH; and C1-4 alkoxy;


each occurrence of Re and Rf is independently selected from the group consisting of: H; C1-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of NR′R″, —OH, halo, C1-4 alkoxy, and C1-4 haloalkoxy; —C(O)(C1-4 alkyl); —C(O)O(C1-4 alkyl); —CONR′R″; —S(O)1-2NR′R″; —S(O)1-2(C1-4 alkyl); —OH; and C1-4 alkoxy;


each occurrence of Rg is independently selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;
    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh; and
    • C6-10 aryl optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;


each occurrence of Rh is independently selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 Ri;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 Ri;
    • heteroaryl of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Ri; and
    • C6-10 aryl optionally substituted with 1-4 Ri;


each occurrence of Ri is independently selected from the group consisting of: C1-6 alkyl; C1-4 haloalkyl; C1-4 alkoxy; C1-4 haloalkoxy; and halo;


each occurrence of Lg is independently selected from the group consisting of: —O—, —NH—, —NRd, —S(O)0-2, C(O), and C1-3 alkylene optionally substituted with 1-3 Ra;


each occurrence of bg is independently 1, 2, or 3; and


each occurrence of R′ and R″ is independently selected from the group consisting of: H; —OH; and C1-4 alkyl.


Variable LA (-(L1)a1-(L2)a2-(L3)a3-(L4)a4-(L5)a5-*, Wherein * Represents the Point of Attachment to Q1)


In some embodiments, LA is a divalent moiety having a 1-6 (e.g., 2-6 (e.g., 2, 3, or 4)) linear array of substituted or unsubstituted carbon and/or heteroatoms. In some embodiments, LA is a divalent moiety having a combination of a cyclic moiety and a 1-6 (e.g., 2-6 (e.g., 2, 3, or 4)) linear array of substituted or unsubstituted carbon and/or heteroatoms. For example, one cyclic moiety (e.g., C3-6, e.g., C4 cycloalkylene), and an acyclic moiety (e.g., O).


In some embodiments, provided that when a3 is 0; and a4 is 1, then L4 is other than straight-chain C1-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb.


In some embodiments, a2 is 1. In some embodiments, a2 is 0.


In certain embodiments (when a2 is 1), L2 is straight-chain C1-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb.


In certain of the foregoing embodiments, L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb.


In certain of the foregoing embodiments, L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


In certain embodiments, L2 is selected from the group consisting of: —CH2—, —CHRb—and —C(Rb)2—. For example, L2 can be —CH2—.


In certain embodiments (when L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb), L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


In certain of these embodiments, L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb. In certain of the foregoing embodiments, L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -(L3)a3-. For example, L2 can be —CH2CH2—.


In certain embodiments, L2 is straight-chain C3 alkylene which is optionally substituted with 1-3 Rb. For example, L2 can be selected from the group consisting of:




embedded image


wherein the asterisk represents point of attachment to -(L3)a3-.


In certain embodiments (when a2 is 1), L2 is straight-chain C2-6 alkenylene, which is optionally substituted with 1-6 Rb. In certain of these embodiments, L2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 Rb. For example, L2 can be selected from the group consisting of:




embedded image


wherein the asterisk represents the point of attachment to -(L3)a3-.


In certain embodiments (when a2 is 1), L2 is selected from the group consisting of:

    • C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc; and
    • heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc.


In certain of these embodiments, L2 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


In certain of the foregoing embodiments, L2 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0, 1, or 2; Q2 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L3)a3-.


In certain of these embodiments, Q2 is CH.


In certain embodiments (when L2 is:




embedded image


as defined supra), n1 and n2 are each 0.


As a non-limiting example (when L2 is:




embedded image


as defined supra), L2 can be




embedded image


wherein the asterisk represents the point of attachment to -(L3)a3- or -(L1)a1, e.g., -(L1)a1, in which at is 1. For example, L2 can be




embedded image


wherein the asterisk represents the point of attachment to -(L1)a1. In certain of these embodiments, -(L1)a1 is 0. In certain of the foregoing embodiments, each of a3, a4, and a5 is 0.


In some embodiments, at is 1. In some embodiments, at is 0.


In certain embodiments (when at is 1), L1 is selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, and —S—. In certain of these embodiments, L1 is —O—.


In some embodiments, a3 is 1. In some embodiments, a3 is 0.


In certain embodiments (when a3 is 1), L3 is selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, and —S—. In certain of these embodiments, L3 is —O—. In certain other embodiments, L3 is —N(H)— or —N(Rd)— (e.g., —N(H)—).


In some embodiments, a4 is 1. In some embodiments, a4 is 0.


In certain embodiments (when a4 is 1), L4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb. In certain of these embodiments, L4 is —CH2—.


In certain embodiments (when a4 is 1), L4 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


In certain of these embodiments, L4 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n3 and n4 are independently 0, 1, or 2; Q3 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L5)a5-.


In certain embodiments (when L4 is:




embedded image


n3 and n4 are each 1. In certain embodiments (when L4 is:




embedded image


Q3 is N.

As anon-limiting example of the foregoing embodiments, L4 can be




embedded image


wherein the asterisk represents the point of attachment to -(L5)a5-.


In some embodiments, a5 is 0.


Non-Limiting Combinations of -(L1)a1-(L2)a2-(L)a3-(L4)a4-(L5)a5-*.


In some embodiments, -(L1)a1-(L2)a2-(L3)a3-(L4)a4-(L5)a5-* has a length of from 1 atom to 8 atoms (as used here and for counting purposes only, moieties such as CH2, C(O), CF2 and the like, whether present in acyclic or cyclic moieties, count as 1 atom); e.g., from 1 atom to 6 atoms, or from 1 atom to 5 atoms, or from 1 atom to 4 atoms; or from 1 atom to 3 atoms; or from 2 atoms to 6 atoms; or from 2 atoms to 4 atoms.


In certain embodiments, one of at, a3, and a5 is 1, and the other two of at, a3, and a5 are 0. In certain embodiments, at is 1, e.g., when L2 is a cyclic group (e.g., cycloalkylene).


In certain embodiments, one of a2 and a4 is 1, and the other of a2 and a4 is 0 or 1.


In certain of the foregoing embodiments,


one of at, a3, and a5 is 1, and the other two of at, a3, and a5 are 0; and


one of a2 and a4 is 1, and the other of a2 and a4 is 0 or 1.


In certain embodiments, 1≤a1+a2+a3+a4+a5≤4. In certain of these embodiments, 1≤a1+a2+a3+a4+a5≤3.


In certain embodiments, a1 and a2 are each 1.


[AA1] In certain embodiments,


a1 and a2 are each 1;


L1 is —O—, —N(H)—, or —N(Rd)—;


L2 is selected from the group consisting of:

    • straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb;
    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


[AA2] In certain embodiments,


a1 and a2 are each 1;


L1 is —O—; and


L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


[AA3] In certain embodiments,


a1 and a2 are each 1;


L1 is —O—; and


L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—.


[AA4] In certain embodiments,


a1 and a2 are each 1;


L1 is —O—; and


L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


In certain embodiments of [AA4], L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb. As non-limiting examples of the foregoing embodiments, L2 can be selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -(L3)a3-. For example, L2 can be —CH2CH2—.


[AA5] In certain embodiments,


a1 and a2 are each 1;


L1 is —O—;


L2 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


In certain embodiments of [AA5], L2 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0, 1, or 2; Q2 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L3)a3-.


In certain of these embodiments, n1 and n2 are independently 0 or 1, optionally 0; and Q2 is CH. For example, n1 and n2 can both be 0; and Q2 can be CH, e.g., L2 can be optionally substituted cyclobutane-diyl, e.g, optionally substituted cyclobutane-1,3-diyl.


In certain embodiments when a1 and a2 are each 1, a3, a4, and a5 are each 0.


In certain embodiments of [AA1], a3, a4, and a5 are each 0. In certain embodiments of [AA2], a3, a4, and a5 are each 0. In certain embodiments of [AA3], a3, a4, and a5 are each 0. In certain embodiments of [AA4], a3, a4, and a5 are each 0. In certain embodiments of [AA5], a3, a4, and a5 are each 0.


In certain embodiments when a1 and a2 are each 1, a3 and a5 are 0; and a4 is 1.


In certain embodiments of [AA1], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA2], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA3], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA4], a3 and a5 are 0; and a4 is 1. In certain embodiments of [AA5], a3 and a5 are 0; and a4 is 1.


In certain embodiments (when a1 and a2 are each 1, a3 and a5 are 0; and a4 is 1), L4 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


In certain of these embodiments, L4 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n3 and n4 are independently 0, 1, or 2; Q3 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L5)a5-. In certain of the foregoing embodiments, n3 and n4 are independently 0 or 1; and Q3 is N.


In certain embodiments, at is 0; and a2 is 1.


[BB1] In certain embodiments, at is 0; a2 is 1; and L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb.


In certain embodiments of [BB1], L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb. In certain of the foregoing embodiments, L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—. For example, L2 can be —CH2—.


In certain embodiments of [BB1], L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb. In certain of the foregoing embodiments, L2 is straight-chain C2 alkylene, which is optionally substituted with 1-3 Rb. As non-limiting examples, L2 can be selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -(L3)a3-. For example, L2 can be —CH2CH2—.


In certain embodiments of [BB1], L2 is straight-chain C3 alkylene, which is optionally substituted with 1-3 Rb. In certain of these embodiments, L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents point of attachment to -(L3)a3-.


In certain embodiments (when at is 0; and a2 is 1), a3 is 0; and a4 is 0.


In certain embodiments of [BB1], a3 is 0; and a4 is 0.


In certain embodiments (when at is 0; and a2 is 1), a3 is 1. In certain embodiments of [BB1], a3 is 1.


In certain embodiments (when at is 0; and a2 is 1) or in certain embodiments of [BB1], a3 is 1; and L3 is selected from the group consisting of: is —O—, —N(H)—, and —N(Rd)—. In certain of these embodiments, a3 is 1; and L3 is —O—. In certain other embodiments, a3 is 1; and L3 is —N(H)— or —N(Rd)—, optionally —N(H)—.


In certain embodiments (when at is 0; and a2 is 1) or in certain embodiments of [BB1], a4 is 1; and L4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb. In certain of these embodiments, a4 is 1; and L4 is —CH2—.


In certain embodiments (when at is 0; and a2 is 1) or in certain embodiments of [BB1], a4 is 0.


In certain embodiments (when at is 0; and a2 is 1) or in certain embodiments of [BB1], a5 is 0.


In certain embodiments (when at is 0; and a2 is 1) or in certain embodiments of [BB1], LA is —CH2—O—CH2—.


[CC1] In certain embodiments, at is 0; a2 is 1; L2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 Rb.


In certain embodiments of [CC1], L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents the point of attachment to -(L3)a3-.


In certain embodiments of [CC1], a3 is 0; and a4 is 0.


For the avoidance of doubt when any one or more of at, a2, a3, a4, and a5 are 0, this means that the corresponding variable (L1-L5) is absent from LA. For example, when each of a3, a4, and a5 are 0, this means that LA has the formula -L1-L2-.


In certain embodiments, LA is -L1-L2-.


In certain embodiments, LA is -L2-L3-.


In certain embodiments, LA is -L2-L3-L4-.


In certain embodiments, LA can be —CH2CH2—O—*, wherein * represents the point of attachment to Q1.


In certain embodiments, LA can be —O—CH2CH2—*, wherein * represents the point of attachment to Q1.


In certain embodiments, LA can be —CH2—O—CH2—.


In certain embodiments, LA can be




embedded image


wherein * represents the point of attachment to Q1.


Variable Q1


In some embodiments, Q1 is selected from the group consisting of:

    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc′; and
    • C6-10 aryl optionally substituted with 1-4 Rc′.


In certain of these embodiments, Q1 is selected from the group consisting of:

    • heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


In certain of the foregoing embodiments, Q1 is selected from the group consisting of:

    • heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


In certain embodiments, Q1 is phenyl optionally substituted with 1-3 Rc′. In certain


of these embodiments, Q1 is selected from the group consisting of:




embedded image


In certain embodiments, Q1 is heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 Rc′In certain of these embodiments, Q1 is pyridyl, which is optionally substituted with 1-3 Rc′In certain of the foregoing embodiments, Q1 is selected from the group consisting of:




embedded image


In certain embodiments, Q1 is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc′.


In certain of these embodiments, Q1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc′.


In certain of the foregoing embodiments, Q1 is heterocyclyl of 4-8 ring atoms, wherein 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, provided that one ring atom is N(Rd),


and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc′.


As non-limiting examples of the foregoing embodiments, Q1 can be




embedded image


wherein m1 and m2 are each independently 0, 1, or 2; and wherein Q1 is optionally substituted with 1-2 Rc′. For example, Q1 can be




embedded image


As another non-limiting example, Q1 can be




embedded image


As another non-limiting example of the foregoing embodiments, Q1 can be




embedded image


(e.g.,




embedded image


optionally substituted with 1-2 Rc′. As another non-limiting example, Q1 can be




embedded image


(e.g.,




embedded image


In certain embodiments, each Rd present in Q1 is independently selected from the group consisting of: —C(O)O(C1-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected Ra.


In certain of the foregoing embodiments, each Rd present in Q1 is C1-6 alkyl optionally substituted with 1-3 independently selected halo.


In certain of the foregoing embodiments, each Rd present in Q1 is C1-4 alkyl substituted with 1-3 —F. In certain embodiments, each Rd present in Q1 is C2-3 alkyl substituted with 1-3 —F. For example, each Rd present in Q1 can be —CH2CF3.


In certain embodiments, each occurrence of Rc′ is an independently selected Rc.


In certain embodiments, each occurrence of Rc′ is independently selected from the group consisting of: (i) C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 independently selected Rc; (ii) heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected Rc; (iii) heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; and (iv) C6-10 aryl optionally substituted with 1-4 Rc.


In certain embodiments, each occurrence of Rc′ is any combination of an independently selected Rc and a cyclic moiety independently selected from the group consisting of: (i) C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 independently selected Rc; (ii) heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected Rc; (iii) heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; and (iv) C6-10 aryl optionally substituted with 1-4 Rc.


In certain embodiment, the cyclic moiety is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected Rc.


In certain embodiment, the cyclic moiety is C6-10 aryl optionally substituted with 1-4 Rc.


In certain embodiments, each Rc present in Q1 is independently selected from the group consisting of: halo; cyano; C1-4 alkoxy; C1-4 haloalkoxy; and C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra.


In certain embodiments, each Rc present in Q1 is independently selected from the group consisting of: halo; cyano; C1-4 alkoxy; C1-4 haloalkoxy; and C1-6 alkyl which is optionally substituted with 1-6 independently selected halo.


In certain of the foregoing embodiments, each Rc present in Q1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo.


In certain embodiments, each Rc present in Q1 is C1-3 alkyl which is optionally substituted with 1-6 —F. For example, each Rc present in Q1 can be CF3.


In certain embodiments, each Rc present in Q1 is an independently selected halo (e.g., —F or —Cl).


Variables Y1, Y2, Y3, X1, and X2


In some embodiments, Y1 is CR1.


In some embodiments, Y2 is CR1.


In some embodiments, Y3 is CR1.


In certain embodiments, each occurrence of R′ is independently H or Rc. In certain of these embodiments, each occurrence of R1 is H.


In certain other embodiments, 1-2 occurrence of R1 is Rc; and each remaining occurrence of R1 is H. For example, one occurrence of R1 can be halo (e.g., —F or —C1); and each remaining occurrence of R1 can be H.


In certain embodiments, Y1, Y2, and Y3 are each independently selected CR1.


In certain embodiments, Y1, Y2, and Y3 are each CH.


In certain embodiments, one of Y1, Y2, and Y3 is CRc, optionally C-halo; and each of the remaining two Y1, Y2, and Y3 is CH.


In some embodiments, X1 is NR2. In certain of these embodiments, X1 is NH.


In some embodiments, X2 is CR5. In certain of these embodiments, X2 is CH.


In certain embodiments, X1 is NR2; and X2 is CR5. In certain of the foregoing embodiments, X1 is NH; and X2 is CH.


In certain embodiments, Y1, Y2, and Y3 are each an independently selected CR1; X1 is NR2; and X2 is CR5. In certain of the foregoing embodiments, Y1, Y2, and Y3 are each CH; X1 is NH; and X2 is CH.


Variables R6 and W


In some embodiments, R6 is H.


[1] In some embodiments, W has formula (A-1):




embedded image


in which:


Ring B1 is a heteroarylene of 5 ring atoms, wherein 1-4 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S; wherein the heteroarylene of Ring B1 is optionally substituted with 1-2 substituents independently selected from the group consisting of oxo and Rc, provided that Ring B1 is attached to the C(═O)NR6 group via a ring carbon atom;


each LAA is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-2 Ra; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa1 is 0, 1, or 2;


Ring C1 is selected from the group consisting of:

    • C3-12 cycloalkylene or C3-12 cycloalkenylene, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and (LAA)aa1-Rg;
    • heterocyclylene or heterocycloalkenylene of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and (LAA)aa1-Rg;
    • heteroarylene of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroarylene is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc and (LAA)aa1-Rg; and
    • C6-10 arylene optionally substituted with 1-4 substituents independently selected from the group consisting of Rc and (LAA)aa1-Rg; R7 is selected from the group consisting of: Rg and -(L7)b7-Rg;


each L7 is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-2 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O); and


b7 is 1, 2, or 3.


The Variable Ring B1

In some embodiments, Ring B1 is a heteroarylene of 5 ring atoms, wherein 1-3 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, O, and S, wherein the heteroarylene of Ring B1 is optionally substituted with 1-2 RcB; and each RcB is an independently selected Rc.


In some embodiments, Ring B1 is a heteroarylene of 5 ring atoms, wherein 2-3 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S, wherein the heteroarylene of Ring B1 is optionally substituted with 1-2 RcB; and each RcB is an independently selected Rc.


In some embodiments, Ring B1 is a heteroarylene of 5 ring atoms, wherein 2-3 of the ring atoms are heteroatoms each independently selected from the group consisting of: N and NH, wherein the heteroarylene of Ring B1 is optionally substituted with 1-2 RcB; and each RcB is an independently selected Rc. As non-limiting examples of the foregoing embodiments, Ring B1 is selected from the group consisting of imidazolylene, pyrazolylene, or triazolylene (such as 1,2,3-triazolylene) which is optionally substituted with one RcB.


In certain embodiments, Ring B1 is imidazolylene, which is optionally substituted with one RcB.


In certain embodiments, Ring B1 is




embedded image


which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is triazolylene (such as 1,2,3-triazolylene) which is optionally substituted with one RcB.


In certain embodiments, Ring B1 is




embedded image


which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is pyrazolylene, which is optionally substituted with one RcB.


In certain embodiments, Ring B1 is




embedded image


or each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, each RcB is independently halo or C1-3 alkyl optionally which is optionally substituted 1-3 independently selected Ra (such as 1-3 independently selected halo).


In some embodiments, Ring B1 is selected from the group consisting of isoxazolylene, oxadiazolylene, oxazolylene, thiazolylene, isothiazolylene, or thiadiazolylene, which is optionally substituted with one RcB.


In certain embodiments, Ring B1 is




embedded image


each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


or each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


or each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, Ring B1 is




embedded image


each of which is optionally substituted with one RcB, wherein aa is the point of connection to (LAA)aa1.


In certain embodiments, each RcB is independently halo or C1-3 alkyl optionally which is optionally substituted 1-3 independently selected Ra (such as 1-3 independently selected halo).


The Variables aa1 and LAA

In some embodiments, aa1 is 0. In some other embodiments, aa1 is 1.


In some embodiments, LAA is C1-3 alkylene optionally substituted with 1-2 Ra1. In certain of these embodiments, LAA is CH2 or CH(Me), such as CH2.


In some embodiments, aa1 is 1; and LAA is C1-3 alkylene optionally substituted with 1-2 Ra1. In certain of these embodiments, LAA is CH2 or CH(Me), such as CH2.


The Variable Ring C1

In some embodiments, Ring C1 is selected from the group consisting of:

    • heteroarylene of 5-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroarylene is optionally substituted with 1-4 substituents independently selected from the group consisting of RcC and RhC; and
    • C6-10 arylene optionally substituted with 1-4 substituents independently selected from the group consisting of RcC and RhC, wherein each RcC is an independently selected Rc; and each RhC is an independently selected Rh.


In certain of these embodiments, Ring C1 is selected from the group consisting of:

    • heteroarylene of 5-6 (such as 6) ring atoms, wherein 1-3 (such as 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroarylene is optionally substituted with 1-4 substituents independently selected from the group consisting of RcC; and
    • C6 arylene optionally substituted with 1-4 substituents independently selected from the group consisting of RcC.


In certain embodiments (when Ring C1 is selected from the group consisting of: heteroarylene of 5-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroarylene is optionally substituted with 1-4 substituents independently selected from the group consisting of RcC and Rhc; and C6-10 arylene optionally substituted with 1-4 substituents independently selected from the group consisting of RcC and Rhc wherein each RcC is an independently selected Rc; and each RhC is an independently selected Rh), Ring C1 is selected from the group consisting of:

    • pyridylene optionally substituted with 1-3 (such as 1) substituents independently selected from the group consisting of RcC; and
    • C6 arylene optionally substituted with 1-4 (such as 1-2) substituents independently selected from the group consisting of RcC.


In certain embodiments, Ring C1 is a group of the following formula:




embedded image


wherein each one of Q1, Q2, Q3, and Q4 is independently selected from the group consisting of N, CH, and CRcC; and bb is the point of connection to R7, wherein each RcC is an independently selected Rc.


In certain embodiments, each one of Q1, Q2, Q3, and Q4 is independently CH or CRcC. In certain other embodiments 1-2 (e.g., 1) of Q1, Q2, Q3, and Q4 are N; and each remaining one of Q1, Q2, Q3, and Q4 are independently CH or CRcC.


In certain of embodiments, Q2 is CH. In certain embodiments, Q3 is CH. In certain embodiments, Q4 is N. In certain embodiments, Q1 is CH. In certain other embodiments, Q1 is CRcC.


In certain embodiments, Ring C1 is




embedded image


such as




embedded image


In certain embodiments, each RcC is independently selected from the group consisting of: -halo and C1-6 (e.g., C1-3) alkyl which is optionally substituted with 1-6 independently selected Ra (e.g., 1-6 independently selected halo, such as —F).


In certain embodiments, each RcC is independently halo, such as —C1 or —F, such as —F.


The Variable R7

In some embodiments, R7 is R9.


In some embodiments, R7 is selected from the group consisting of:

    • C3-12 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc7, Rh7, and -(Lg)bg-Rh7; and
    • heterocyclyl of 4-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc7, Rh7, and -(Lg)bg-Rh7, wherein each Rc7 is an independently selected Rc; and Rh7 is an independently selected Rh.


In certain of these embodiments, R7 is selected from the group consisting of:

    • C4-8 (e.g., C4, C5, or C6) cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc7, and Rh7; and
    • heterocyclyl of 4-8 (e.g., 4, 5, or 6) ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc7, and Rh7.


In certain of the foregoing embodiments, R7 is selected from the group consisting of:

    • C6 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc7; and
    • heterocyclyl of 6 ring atoms, wherein 1-2 (such as one) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc7.


In certain of these embodiments, R7 is a group of the following formula:




embedded image


wherein X7 is CH, CRc7, or N, such as CH or N. In certain embodiments (when R7 is




embedded image


two Rc7 groups are present.


In certain embodiments, R7 is a group of the following formula:




embedded image


wherein X7 is N or CH; and each Rc7 is an independently selected Rc. In certain embodiments, R7 is




embedded image


wherein X7 is N or CH; such as




embedded image


In certain of the foregoing embodiments, R7 is selected from the group consisting of:

    • C4 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc7; and
    • heterocyclyl of 4 ring atoms, wherein 1-2 (such as one) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc7.


In certain of these embodiments, R7 is a group of the following formula:




embedded image


wherein X7 is CH, CRc7, or N, such as CH or N. In certain embodiments (when R7 is




embedded image


two Rc7 groups are present.


In certain embodiments, R7 is a group of the following formula:




embedded image


wherein X7 is N or CH; and each Rc7 is an independently selected Rc. In certain embodiments, R7

is




embedded image


wherein X7 is N or CH; such as




embedded image


In certain embodiments, R7 is selected the group consisting of tetrahydropyranyl, morpholinyl, 5-azaspiro[2.5]octanyl, or 2-azabicyclo[2.2.1]heptanyl, each of which is optionally substituted with 1-2 R7. For example, R7 can be:




embedded image


In certain embodiments, each Rc7 is an independently selected halo or C1-3 alkyl optionally substituted with 1-6 Ra (e.g., 1-6 independently selected halo). In certain of these embodiments, each Rc7 is independently halo, such as —F.


In some embodiments, R7 is selected from the group consisting of:

    • C4-5 cycloalkyl, which is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc7; and
    • heterocyclyl of 5-6 ring atoms, wherein 1-2 (such as one) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc7.


In certain embodiments, R7 is a group of the following formula:




embedded image


wherein X7 is CH, CRc7, or N, such as CH or N.


In certain embodiments, R7 is a group of the following formula:




embedded image


wherein Rd is independently selected from the group consisting of: C1-6 alkyl optionally substituted with 1-3 independently selected Ra.


In certain embodiments, R7 is selected from the group consisting of tetrahydropyranyl, morpholinyl, 5-azaspiro[2.5]octanyl, or 2-azabicyclo[2.2.1]heptanyl, each of which is optionally substituted with 1-2 Rc7. For example, R7 can be:




embedded image


[2] In some embodiments, W has formula (A-2):




embedded image


in which:


Ring B2 is a heteroarylene of 5 ring atoms, wherein 1-4 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S, wherein the heteroarylene of Ring B is optionally substituted with 1-2 substituents independently selected from the group consisting of: oxo and Rc, provided that Ring B is attached to the C(═O)NR6 group via a ring carbon atom;


each LAB is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa2 is 0, 1, 2, or 3;


Ring C2 is selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;
    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; and
    • C6-10 aryl optionally substituted with 1-4 Rc;


The Variable Ring B2

In some embodiments, Ring B2 is a heteroarylene of 5 ring atoms, wherein from 2-3 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S, wherein the heteroarylene of Ring B2 is optionally substituted with from 1-2 Rc, provided that Ring B2 is attached to the C(═O)NR6 group via a ring carbon atom.


In certain of these embodiments, Ring B2 is selected from the group consisting of: pyrazolylene; imidazolylene; thiazolylene; oxazolylene; triazolylene such as 1,2,3-triazolylene or 1,2,4-triazolylene; isoxazolylene; and isothiazolylene, each of which is optionally substituted with Rc; and a ring nitrogen is optionally substituted with Rd.


As non-limiting examples of the foregoing embodiments, Ring B2 can be pyrazolylene; imidazolylene; 1,2,3-triazolylene; 1,2,4-triazolylene, each of which is optionally substituted with Rc; and a ring nitrogen is optionally substituted with Rd.


In some embodiments, Ring B2 has Formula B1a or B2a:




embedded image


B4 is C or N;


B1, B2, and B3 are each independently CH, CRc, NH, N(Rd), N, O, or S; provided that from 0-2 of B1, B2, and B3 is CRc;


aa is the point of attachment to (LA)a1; and


each custom-character is independently a single bond or a double bond provided that the ring including B1-B4 is a heteroaryl.


In certain embodiments, Ring B2 has Formula B1a.


In certain embodiments of (B1a), B4 is N.


In certain embodiments, Ring B2 is:




embedded image


For example, Ring B2 can be




embedded image


In certain embodiments, Ring B2 is




embedded image


wherein B1 and B2 are independently CH, CRc, NH, N(Rd), N, O, or S.


In certain of these embodiments, Ring B2 is




embedded image


In certain embodiments, Ring B2 is




embedded image


wherein B2 and B3 are independently CH, CRc, or N.


In certain of these embodiments, Ring B2 is




embedded image


As a non-limiting example of the foregoing embodiments, Ring B2 can be




embedded image


As further non-limiting examples, Ring B2 can be or




embedded image


For example, Ring B2 can be




embedded image


As further non-limiting examples, Ring B2 can be




embedded image


For example, Ring B2 can be




embedded image


In certain embodiments of (B1a), B4 is C.


In certain embodiments, Ring B2 is




embedded image


wherein one of B1 and B2 is NH, NRd, O, or S; and the other one of B1 and B2 is N. In certain of these embodiments, B3 is CH or CRc, such as CH. As non-limiting examples of these embodiments, Ring B2 can be




embedded image


each of which is further optionally substituted with Rc (such as not further optionally substituted).


In certain embodiments, Ring B2 is




embedded image


wherein one of B1 and B3 is NH, NRd, O, or S; and the other one of B1 and B3 is N, wherein Ring B2 is further optionally substituted with Rc.


In certain embodiments, Ring B2 is




embedded image


wherein one of B2 and B3 is NH, NRd, O, or S; and the other one of B2 or B3 is N, wherein Ring B2 is further optionally substituted with Rc.


As non-limiting examples, Ring B2 can be




embedded image


or each of which is optionally substituted with Rc (such as unsubstituted).


In certain embodiments, Ring B2 has Formula (B2a). In certain embodiments of (B2a), B4 is N. As non-limiting examples of the foregoing embodiments, Ring B2 is




embedded image


each of which is optionally substituted with Rc (such as unsubstituted).


In certain embodiments, each Rc substituent of Ring B2 is independently —OH; C1-3 alkyl; C1-3 alkyl optionally substituted with from 1-6 independently selected halo; halo; cyano; C1-4 alkoxy; or C1-4 haloalkoxy.


The Variables LAB and aa2


In some embodiments, a1 is 0. In some embodiments, a1 is 1.


In some embodiments, LAB is C1-3 alkylene optionally substituted with from 1-4 Ra1. In certain of these embodiments, LAB is CH2 optionally substituted with from 1-2 Ra1 In certain embodiments, LAB is C(H)Me optionally substituted with from 1-4 Ra1, such as wherein LAB is C(H)Me. In certain embodiments, LAB is CH2CH2.


In certain embodiments, aa2 is 1; and LAB is C1-3 alkylene optionally substituted with from 1-4 Ra1. In certain of these embodiments, LA is CH2 optionally substituted with from 1-2 Ra1. In certain embodiments, LAB is C(H)Me optionally substituted with from 1-4 Ra1, such as wherein LA is C(H)Me. In certain embodiments, LAB is CH2CH2.


In certain embodiments, aa2 is 2; and (LAB)aa2 is -LA1-LA2, wherein LA1 and LA2 are independently selected LA, and LA2 is the point of attachment to Ring C2. In certain of these embodiments, LA1 is C1-3 alkylene optionally substituted with from 1-4 Ra1, such as CH2, C(H)Me, or CH2CH2. In certain of the foregoing embodiments, LA2 is —O—.


The Variable Ring C2

In some embodiments, Ring C2 is selected from the group consisting of:

    • heteroaryl of 5-12 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(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
    • C6-10 aryl optionally substituted with from 1-4 Rc.


In some embodiments, Ring C2 is selected from the group consisting of:

    • heteroaryl of 5-6 ring atoms, wherein from 1-3 (such as 1-2) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
    • phenyl optionally substituted with from 1-4 Rc.


In certain embodiments, Ring C2 is




embedded image


wherein Q1, Q2, Q3, Q4, and Qs are independently CH, CRc, or N, provided that at least two of Q1-Q5 are CH.


In certain of these embodiments, Q3 is CRc. In certain embodiments, each one of Q1, Q2, Q4, and Q5 is independently CH or CRc. As non-limiting examples of the foregoing embodiments, Ring C2 can be:




embedded image


such as




embedded image


In certain embodiments, Rc is C1-C4 haloalkyl (e.g., fluoroalkyl or perfluoroalkyl), e.g., C1-C2 haloalkyl (e.g., fluoroalkyl or perfluoroalkyl), e.g., C1 haloalkyl (e.g., fluoroalkyl or perfluoroalkyl), e.g., CF3.


In certain embodiments, one of Q1 and Q2 is N; and each remaining one of Q1, Q2, Q4, and Q5 is independently CH or CRc. As non-limiting examples of the foregoing embodiments, Ring C2 is




embedded image


In certain embodiments, Q2 is CRC. In certain of these embodiments, each one of Q1, Q3, Q4, and Q5 is independently CH or CRc. As non-limiting examples of the foregoing embodiments, Ring C2 can be




embedded image


such as




embedded image


In certain embodiments, Q2 is CRc; one of Q1 and Q3 (such as Q1) is N; and each remaining one of Q1, Q3, Q4, and Q5 is independently CH or CRc. In certain of these embodiments, each one of Q1, Q2, Q3, Q4, and Q5 is CH (i.e., Ring C2 is unsubstituted phenyl).


In certain embodiments, one of Q1 and Q2 is N; and each remaining one of Q1, Q2, Q3, Q4, and Q5 is CH, such as wherein Ring C2 is




embedded image


or wherein Ring C2 is




embedded image


In some embodiments, Ring C2 is selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, and
    • heterocyclyl or heterocycloalkenyl of 3-12 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(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.


In certain of these embodiments, Ring C2 is selected from the group consisting of:

    • C3-6 cycloalkyl optionally substituted with from 1-4 Rc, and
    • heterocyclyl of 4-6 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with from 1-4 Rc.


In certain embodiments, Ring C2 is C3-6 cycloalkyl optionally substituted with from 1-2 Rc, such as wherein Ring C2 is cyclohexyl; or wherein R6 is cyclohexyl substituted with from 1-2 Rc (e.g., halo).


In certain embodiments, each Rc substituent of Ring C2 is selected from the group consisting of: halo; cyano; C1-6 alkyl; C1-6 alkyl substituted with from 1-6 Ra; C1-4 alkoxy; and C1-4 haloalkoxy. In certain of these embodiments, one occurrence of Rc substituent of Ring C is C1-6 alkyl or C1-6 alkyl substituted with from 1-6 Ra, such as C1-6 alkyl substituted with from 1-6 independently selected halo, such as —F.


[3] In some embodiments, W is heteroaryl of 5 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; provided the heteroaryl is attached to the C(═O)NR6 group via a ring carbon atom.


In certain embodiments, W is thienyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl, or tetrazolyl, e.g., thiazolyl.


In certain of the foregoing embodiments, the heteroaryl of 5 ring atoms is unsubstituted.


In certain of the foregoing embodiments, the heteroaryl of 5 ring atoms is optionally substituted with 1-4 (e.g., 1-3, 1-2, or 1) Rc.


[4] In some embodiments, W has formula (A-3):




embedded image


in which


P1, P2, P3, P4, and P5 are each independently selected from the group consisting of: N, NH, NRd, NR71, CH, CRc, CR71, and C(═O), provided that 1-3, such as 1, of P2, P3, and P4 is CR71 or NR71;


each occurrence of R71 is independently -(LAC)aa3-R8, wherein:


each LAC is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra; —O—; —NRN; —S(O)0-2; C(O); C(O)O; OC(O); NRNC(O); C(O)NRN; NRNC(O)NRN; NRNC(O)O; and OC(O)NRN;

    • aa3 is 0, 1, 2, or 3;
    • each occurrence of R8 is independently Rg or C1-10 alkyl optionally substituted with 1-6 Ra1; and


each occurrence of RN is independently H or Rd.


In some embodiments, W has formula (A-3-1)




embedded image


P1, P2, P3, P4, and P5 are each independently selected from the group consisting of: N, NH, NRd, NR71, CH, CRC, CR71, and C(═O);


each occurrence of R71 is independently -(LAC)aa3-R8, wherein:


each LAC is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra; —O—; —NRN; —S(O)0-2; C(O); C(O)O; OC(O); NRNC(O); C(O)NRN; NRNC(O)NRN; NRNC(O)O; and OC(O)NRN;


aa3 is 0, 1, 2, or 3;


each occurrence of R8 is independently Rg or C1-10 alkyl optionally substituted with 1-6 Ra1; and


each occurrence of RN is independently H or Rd;


The Variables P1, P2, P3, P4, and P5


In some embodiments, P1 and P5 are independently CH or CRC; and P2, P3, and P4 are independently CH, CRc, or CR7.


In some embodiments, one of P, P2, P3, P4 and P5 is N. In some embodiments, two of P, P2, P3, P4, and P5 are N.


In some embodiments, one of P2, P3, and P4 is CR71.


In certain embodiments, P3 is CR71. In certain of these embodiments, P4 is N. In certain other embodiments, P4 is CH or CRc. In certain of the foregoing embodiments, P1 is N. In certain other embodiments, P1 is CH or CRc. In certain embodiments, P2 and P5 are independently CH or CRc.


In certain embodiments, P3 is CR7; P1, P2, P4, and P5 are independently CH or CRc.


In certain embodiments, the




embedded image


moiety has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc7 is an independently selected Rc, such as:




embedded image


In certain embodiments, P3 is CR7, P4 is N; and P1, P2, and P5 are independently CH or CRc.


In certain embodiments, the




embedded image


moiety has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc7 is an independently selected Rc, such as:




embedded image


In certain embodiments, P3 is CR7; P4 and P1 are N; and P2 and P5 are independently CH or CRc.


In certain embodiments, the




embedded image


moiety has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc7 is an independently selected Rc, such as:




embedded image


In some embodiments, P4 is CR71. In certain embodiments, P3 is N. In certain other embodiments, P3 is CH or CRc. In certain embodiments, P, P2, and P5 are independently CH or CRc.


In certain embodiments, P4 is CR71; P3 is CH or CRC; and P1, P2, and P5 are independently CH or CRc.


In certain embodiments, the




embedded image


moiety has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc7 is an independently selected Rc, such as:




embedded image


In certain embodiments, P4 is CR71; P3 is N; and P1, P2, and P5 are independently CH or CRc.


In certain embodiments, the




embedded image


moiety has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc71 is an independently selected Rc, such as:




embedded image


In certain embodiments, the




embedded image


moiety has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc7 is an independently selected Rc.


In certain embodiments, each occurrence of R7 is independently selected from the group consisting of halo; cyano; C1-3 alkyl; C1-4 alkoxy; C1-4 haloalkoxy; and C1-3 alkyl substituted with from 1-6 independently selected halo, such as —F.


In certain embodiments, the




embedded image


moiety is selected from the group consisting of




embedded image


The Variables LAC, aa3, and R′


In some embodiments, aa3 is 0. In some embodiments, aa3 is 1. In some embodiments, aa3 is 2. In some embodiments, aa3 is 3.


In some embodiments, LAC is —O—, —NH—, or —CH2—, such as wherein LAC is —O—. In certain embodiment (when aa3 is 1), LAC is —O—, —NH—, or —CH2—, such as wherein LAC is —O—. In certain embodiments, aa3 is 1; and LAC is —O—.


In certain embodiments, aa31 is 2; and -(LAC)aa3- is -LA1-LA2, wherein LA1 and LA2 are independently selected LAC; and LA2 is the point of attachment to R8. In certain of these embodiments, LA1 is —O—; and LA2 is C1-3 alkylene optionally substituted with from 1-2 Ra, such as wherein LA1 is —O—; and LA2 is CH2.


In certain embodiments, aa3 is 3; and -(LAC)aa3- is -LA1-LA2-LA3, wherein LA1, LA2, and LA3 are independently selected LAC; and LA3 is the point of attachment to R8. In certain of these embodiments, LA1 and LA3 are each independently C1-3 alkylene optionally substituted with from 1-2 Ra. In certain embodiments, LA2 is NRNC(O)O or OC(O)NRNIn some embodiments, R8 is C1-10 alkyl optionally substituted with 1-4 Ra.


In certain embodiments, R8 is C1-10 alkyl, such as C1-7 alkyl, such as C1, C2, C3, C4, C5, C6, or C7 alkyl, such as ethyl or isopropyl.


In certain embodiments, R8 is C1-10 alkyl substituted with 1-6 Ra, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 Ra. In certain embodiments, R1 is selected from the group consisting of: halo, such as —F; —OH; C1-4 alkoxy; and C1-4 haloalkoxy.


In certain embodiments, R8 is C1-10 alkyl substituted with 1-6 independently selected halo, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 independently selected halo. As non-limiting examples of the foregoing embodiments, R8 is C1-10 alkyl substituted with 1-6 —F, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 —F, such as




embedded image


In certain embodiments, R8 is C1-10 alkyl substituted with —OH, C1-4 alkoxy, or C1-4 haloalkoxy, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 independently selected C1-4 alkoxy, such as




embedded image


In some embodiments, R8 is R9.


In certain embodiments, R8 is selected from the group consisting of:

    • C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, Rh, and -(Lg)bg-Rh; and
    • heterocyclyl or heterocycloalkenyl of 4-8 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(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, Rh, and -(L)bg-Rh.


In certain embodiments, R8 is selected from the group consisting of:

    • C3-8 cycloalkyl which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
    • heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.


In certain embodiments, R8 is selected from the group consisting of:

    • C3-8 cycloalkyl substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc; and
    • heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc.


As non-limiting examples of the foregoing embodiments, R8 can be selected from the group consisting of piperidinyl, pyrrolidinyl, azetidinyl, azaspiro[3.3]heptanyl, cyclobutyl, cyclopentyl, and cyclohexyl, each of which is substituted with 2 —F and further optionally substituted with from 1-2 Rc, such as




embedded image


As another non-limiting example, R8 can be optionally substituted 3-azabicyclo[3.1.0]hexane, e.g.:




embedded image


In certain embodiments, R8 is selected from the group consisting of:

    • C3-8 cycloalkyl such as cyclopropyl, cyclohexyl, cyclobutyl, or cyclopentyl;
    • C3-8 cycloalkyl substituted with a substituent selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-4 alkoxy substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkyl; and C1-6 alkyl substituted from 1-6 independently selected halo, C1-4 alkoxy, or C1-4 haloalkoxy, wherein the cycloalkyl is further optionally substituted with from 1-2 Rc;
    • heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, such as:




embedded image


and

    • heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is substituted with a substituent selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-4 alkoxy substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkyl; and C1-6 alkyl substituted from 1-6 independently selected halo, C1-4 alkoxy, or C1-4 haloalkoxy, wherein the heterocyclyl is further optionally substituted with from 1-2 Rc, such as




embedded image


In certain embodiments, R8 is C3-8 cycloalkyl such as cyclopropyl, cyclohexyl, cyclobutyl, or cyclopentyl; In certain embodiments, R8 is C3-8 cycloalkyl substituted with a substituent selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-4 alkoxy substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkyl; and C1-6 alkyl substituted from 1-6 independently selected halo, C1-4 alkoxy, or C1-4 haloalkoxy, wherein the cycloalkyl is further optionally substituted with from 1-2 Rc;


In certain embodiments, R8 is heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, such as:




embedded image


In certain embodiments, R8 is heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is substituted with a substituent selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-4 alkoxy substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkyl; and C1-6 alkyl substituted from 1-6 independently selected halo, C1-4 alkoxy, or C1-4 haloalkoxy, wherein the heterocyclyl is further optionally substituted with from 1-2 Rc, such as




embedded image


In certain embodiments, R8 is selected from the group consisting of:

    • heteroaryl of 5-6 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(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
    • C6-10 aryl, such as phenyl, optionally substituted with from 1-4 Rc.


In certain embodiments, aa3 is 0; and R8 is selected from the group consisting of:

    • C3-8 cycloalkyl substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc; and
    • heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc.


As non-limiting examples of the foregoing embodiments, R8 is selected from the group consisting of piperidinyl, pyrrolidinyl, azetidinyl, azaspiro[3.3]heptanyl, cyclobutyl, cyclopentyl, and cyclohexyl, each of which is substituted with 2 —F and further optionally substituted with from 1-2 Rc, such as




embedded image


As another non-limiting example, R8 can be optionally substituted 3-azabicyclo[3.1.0]hexane, e.g.:




embedded image


In certain embodiments, aa3 is 0; and R8 is C1-10 alkyl substituted with 1-6 independently selected halo, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 independently selected halo. As non-limiting examples of the foregoing embodiments, R8 can be C1-10 alkyl substituted with 1-6 —F, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 —F, such as




embedded image


In certain embodiments, aa3 is 1; LAC is —O— or —NH—; and R8 is C1-10 alkyl substituted with 1-6 independently selected halo, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 independently selected halo. In certain of these embodiments, R′ is C1-10 alkyl substituted with 1-6 —F, such as C1, C2, C3, C4, C5, C6, or C7 alkyl substituted with from 1-6 —F, such as




embedded image


In certain embodiments, LA is —O—.


In certain embodiments, aa3 is 1; LAC is —O—, —NH—, or —CH2—; and R8 is selected from the group consisting of:

    • C3-8 cycloalkyl substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc; and
    • heterocyclyl of 4-8 ring atoms, wherein from 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc.


In certain of these embodiments, R8 is C3-8 cycloalkyl substituted with from 1-2 (such as 2) independently selected halo (such as —F) and further optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc, such as cyclobutyl, cyclopentyl, and cyclohexyl, each of which is substituted with 2 —F and further optionally substituted with from 1-2 Rc, such as




embedded image


[5] In certain of the foregoing embodiments, LAC is —O—.


In some embodiments, W is a bicyclic or polycyclic ring system selected from the group consisting of:

    • bicyclic or polycyclic C5-15 cycloalkyl or C5-15 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heterocyclyl or heterocycloalkenyl of 7-15 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heteroaryl of 8-15 ring atoms, wherein 1-6 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of: oxo, Rc, and -(LAD)bB-Rg; and
    • bicyclic or polycyclic C8-15 aryl optionally substituted with 1-4 substituents independently selected from the group consisting of: oxo, Rc, and -(LAD)bB-Rg,
    • provided the bicyclic or polycyclic heteroring is attached to the C(═O)NR6 group via a ring carbon atom;
    • each occurrence of LAD is selected from the group consisting of: —O—, —NH—, —NRd, —S(O)0-2, C(O), and C1-3 alkylene optionally substituted with 1-3 Ra; and
    • bB is 0, 1, 2, or 3.


In some embodiments, W is selected from the group consisting of:

    • bicyclic or polycyclic heteroaryl of 8-15 (e.g., 9, 10, 11, or 12) ring atoms, wherein from 1-6 (e.g., 1, 2-3, or 3-4) ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 substituents independently selected from the group consisting of: Rc and -(LB)bB-Rh; and
    • bicyclic or polycyclic C8-15 (e.g., C9, C10, C11, or C12) aryl optionally substituted with from 1-4 substituents independently selected from the group consisting of: Rc and -(LB)bB-Rh, provided that W is attached to the C(═O)NR6 group via a ring carbon atom.


In some embodiments, W has Formula (B-1):




embedded image


wherein T3 and T4 are independently C or N;


T5, T6, T7, and T8 are independently N, CH, or CRB;


T1 and T2 are independently N, NH, NRd, NRB, CH, CRB, O, or S;


each occurrence of RB is independently Rc or -(LB)bB-Rh; and


each custom-character is independently a single bond or a double bond, provided that the 5-membered ring including T1-T4 is heteroaryl, and the 6-membered ring including T3-T8 is aryl or heteroaryl,


further provided that no more than 4 of T1-T8 are heteroatoms; and no more than 4 RB groups are present.


In certain of these embodiments, the 5-membered ring including T1-T4 is thiophene, thiazole, oxazole, imidazole, or pyrazole.


In certain embodiments, W is selected from the group consisting of:




embedded image


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh, and m1 is 0, 1, or 2 (such as 1 or 2). As non-limiting


examples of the foregoing embodiments, W can be




embedded image


In some embodiments, W has Formula (B-2):




embedded image


wherein T3 and T4 are independently C or N;


T1 and T2 are independently N, NH, NRd, NRB, CH, CRB, O, or S;


T9 is —O—, S(O)0-2, CH2, CHRB, C(RB)2, NH, NRd, or NRB;


nB is 0, 1, 2, or 3; mB is 1 or 2;


each RB is independently Rc or -(LB)bB-Rh.


m1 is 0, 1, or, 2 (such as 1 or 2); and


each custom-character is independently a single bond or a double bond, provided that the 5-membered ring including T1-T4 is heteroaryl, further provided that no more than 4 RB groups are present.


In certain of these embodiments, T3 is N. In certain embodiments, the 5-membered ring including T1-T4 is pyrazole or imidazole.


In certain embodiments, W is




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; m1 is 0, 1, or 2 (such as 1 or 2).


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB, is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or 2 (such as 1 or 2).


In certain embodiments, W is




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or 2 (such as 1 or 2).


In certain embodiments, W is




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or 2 (such as 1 or 2).


In some embodiments, W has Formula (B-3):




embedded image


wherein P3 and P4 are independently C or N;


P1 and P2 are independently N, NH, NRd, NRB, CH, CRB, O, or S;


P5, P6, P7, and P8 are independently N, CH, or CRB;


each RB is independently Rc or -(LB)bB-Rh; and


each custom-character is independently a single bond or a double bond, provided that the 5-membered ring including P1-P4 is heteroaryl, and the 6-membered ring including P3—P8 is aryl or heteroaryl, further provided that no more than 4 of P1-P8 are heteroatoms; and no more than 4 RB groups are present.


In certain of these embodiments, P3 is C. In certain embodiments, P4 is C. In certain of the foregoing embodiments, P3 is C; and P4 is C. In certain embodiments, P1 is N; and P2 is NRB (e.g., N-(LB)bB-Rh). In certain embodiments, P1 is N; and P2 is NH. In certain embodiments, each one of P5, P6, P7, and P8 is independently N, CH, or CRB. For example, P6 is CRB (e.g., CRc); and P5, P7, and P8 are CH.


In certain of embodiments of (B-3), the 5-membered ring including P1-P4 is pyrazole.


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or 2 (such as 1 or 2).


In certain embodiments, W is selected from the group consisting of:




embedded image


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; m1 is 0, 1, or, 2 (such as 0); and each RhB is an independently selected Rh, such as wherein W is




embedded image


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or, 2 (such as 0).


As non-limiting examples of the foregoing embodiments, W can be




embedded image


In certain embodiments of (B3), the 5-membered ring including P1-P4 is imidazole.


In certain of these embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or 2 (such as 0).


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or, 2 (such as


0), such as




embedded image


In some embodiments, W has Formula (B4):




embedded image


wherein P3 and P4 are independently C or N;


P1 and P2 are independently N, NH, NRd, NRB, CH, CRB, O, or S;


Q9 is —O—, S(O)0-2, CH2, CHRB, C(RB)2, NH, NRd, or NRB;


nB is 0, 1, 2, or 3; mB is 1 or 2;


each occurrence of RB is independently Rc or -(LB)bB-Rh.


m1 is 0, 1, or, 2 (such as 1 or 2); and


each custom-character is independently a single bond or a double bond, provided that the 5-membered ring including P1-P4 is heteroaryl, provided that no more than 4 RB groups are present.


In certain of these embodiments, the 5-membered ring including P1-P4 is pyrazole, thiophene, or imidazole.


In certain embodiments, W is selected from the group consisting of:




embedded image


wherein each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or, 2 (such as 0).


In some embodiments, W has Formula (B5) or (B6):




embedded image


wherein BA is a ring of 5-8 ring atoms wherein from 0-3 ring atoms are heteroatoms each independently selected from the group consisting of N, NH, NRd, O, and S, wherein BA is optionally substituted with from 1-2 RB; and


each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or 2 (such as 0).


In certain of these embodiments, W is a non-aromatic ring of 5-8 ring atoms wherein from 0-3 ring atoms are heteroatoms each independently selected from the group consisting of N, NH, NRd, O, and S, wherein BA is optionally substituted with from 1-2 RB.


As non-limiting examples of the foregoing embodiments, W can be selected from the group consisting of:




embedded image


wherein RB is independently Rc or -(LB)bB-Rh.


In certain embodiments of (B5) or (B6), BA is a 5-membered heteroaromatic ring, wherein ring 1-2 ring atoms are heteroatoms each independently selected from the group consisting of N, NH, NRd, O, and S, wherein BA is optionally substituted with from 1-2 RB, such as wherein BA is pyrazole optionally substituted with RB. In certain of these embodiments, Ring B is




embedded image


wherein RhB is an independently selected Rh.


In some embodiments, W has Formula (B7):




embedded image


wherein BB is an aromatic ring of 5-6 ring atoms wherein from 0-3 ring atoms are heteroatoms each independently selected from the group consisting of N, NH, NRd, O, and S, wherein BB is optionally substituted with from 1-2 RB;


each RB is independently Rc or -(LB)bB-Rh; and m1 is 0, 1, or, 2 (such as 0), such as, wherein Ring B is




embedded image


In some embodiments, W is a spirocyclic ring (e.g., [4.4.1], [5.4.1], or [5.5.1] spirocycle). As non-limiting examples, Ring B can be:




embedded image


In some embodiments, W is a bridged ring. As a non-limiting example, Ring B can be:




embedded image


In certain embodiments, each RB is independently selected from the group consisting of: halo; cyano; C1-4 alkyl such as methyl; C1-4 alkyl substituted with from 1-6 independently selected halo, such as —CF3 or —CH2CH2CF3; C1-4 alkoxy, such as methoxy, ethoxy, or isopropoxy; and C1-4 haloalkoxy, such as —OCF3, —OCHF2, or —OCH2CF3.


In certain embodiments, LB is CH2.


In certain embodiments, each Rh substituent of Ring B, such as RhB, is independently selected from the group consisting of:

    • heteroaryl of 5-6 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(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-3 Ri; and
    • C6 aryl optionally substituted with from 1-2 Ri, such as wherein Rg1 is phenyl optionally substituted with from 1-4 Ri.


[6] In some embodiments, W has formula (A-4):




embedded image


LAE is selected from the group consisting of:

    • C1-6 alkylene, C2-6 alkenylene, or C2-6 alkynylene, each of which is optionally substituted with 1-6 Ra;
    • monocyclic C3-8 cycloalkylene or C3-8 cycloalkenylene, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; and
    • monocyclic heterocyclylene or heterocycloalkenylene of 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc, provided that the heterocycloylene or heterocycloalkenylene is attached to the C(═O)NR6 group via a ring carbon atom;


each LAF is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa4 is 0, 1, 2, or 3; and


Ring C4 is R9.


Variable LAE

In some embodiments, LAE is C1-6 alkylene, C2-6 alkenylene, or C2-6 alkynylene, each of which is optionally substituted with from 1-6 Ra1.


In certain embodiments, LAE is C1-6 alkylene optionally substituted with from 1-6 Ra1. In certain embodiments, LAE is CH2. In certain embodiments, LB is branched C2-6 alkylene optionally substituted with from 1-6 Ra1, such as —CH(Me)—, —C(Me)2—, or —C(Me)2—CH2—. In certain embodiments, LAE is linear C2-6 alkylene optionally substituted with from 1-6 Ra1, such as CH2CH2 or CH2CH2CH2.


In certain embodiments, LAE is C2-6 alkenylene optionally substituted with from 1-6 Ra1. In certain of these embodiments, LAE is C2-4 alkenyl optionally substituted with from 1-6 Ra1. In certain embodiments, the NR6C(═O) group and the (LAA)a1 group are attached to two sp2 hybridized carbons of LAE. As non-limiting examples, LAE can be CH═CH or C(Me)=CH*, wherein the asterisk represents point of attachment to (LAF)aa4.


In some embodiments, LAE is selected from the group consisting of:

    • monocyclic C3-8 cycloalkylene or C3-8 cycloalkenylene, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; and
    • monocyclic heterocyclylene or heterocycloalkenylene of 3-8 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(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, provided that the heterocycloylene or heterocycloalkenylene is attached to the C(═O)NR6 group via a ring carbon atom.


In certain embodiments, LAE is monocyclic C3-8 cycloalkylene which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc, such as wherein LAE is C4-8 cycloalkylene which is optionally substituted with from 1-4 Rc, such as wherein LAE is cyclobutylene.


In certain embodiments, LAE is monocyclic heterocyclylene of 4-8 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(O)0-2, and wherein the heterocyclylene is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, such as wherein LB is pyrrolidinylene or morpholinylene, each optionally substituted with oxo and further optionally substituted with from 1-2 Rc, such as wherein LAE is




embedded image


or wherein bb is the point of attachment to (LAF)aa4.


The Variables LAF and aa4

In some embodiments, aa4 is 0.


In some embodiments, aa4 is 1.


In some embodiments, LAF is —O—, —S(O)2—, C(═O), or CH2. In certain embodiments, LAF is —O—. In certain embodiments, LAF is —S(O)2—. In certain embodiments, LAF is C(O). In certain embodiments, LA is CH2.


In some embodiments, aa4 is 1; and LAF is —O—, —S(O)2—, C(═O), or CH2. In certain of these embodiments, LAF is —O—. In certain embodiments, LAF is —S(O)2—. In certain embodiments, LAF is C(═O). In certain embodiments, LAF is CH2.


In some embodiments, aa4 is 2 or 3. In certain of these embodiments, each occurrence of LAF is independently C(═O), S(O)2, NH, N(C1-3 alkyl), —O—, or CH2, provided that (LAF)aa4 does not comprise an 0-0 or N-0 bond.


The Variable Ring C4

In some embodiments, Ring C4 is selected from the group consisting of:

    • heteroaryl of 5-12 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(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 substituents independently selected from the group consisting of Rc, Rh, and -(Lg)bg-Rh; and
    • C6-10 aryl optionally substituted with from 1-4 substituents independently selected from the group consisting of Rc, Rh, and -(Lg)bg-Rh.


In certain of these embodiments, Ring C4 is selected from the group consisting of:

    • heteroaryl of 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(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 RcC; and
    • C6-10 aryl optionally substituted with from 1-4 RcC, wherein each RcC is an independently selected Rc.


In certain embodiments, Ring C4 is selected from the group consisting of:

    • heteroaryl of 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(O)0-2, and wherein the heteroaryl is optionally substituted with from 1-4 RcC; and
    • C6 aryl optionally substituted with from 1-4 RcC, wherein each RcC is an independently selected Rc.


In certain embodiments, Ring C4 is




embedded image


wherein nc is 0 or 1, such as 0; and each RcC is an independently selected Rc.


In certain embodiments, Ring C4 is




embedded image


or wherein Ring C4 is




embedded image


wherein nc is 0 or 1, such as 0; and each RcC is an independently selected Rc.


In some embodiments, Ring C4 is unsubstituted phenyl or pyridyl.


In some embodiments, Ring C4 is selected from the group consisting of:

    • heteroaryl of 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(O)0-2, and wherein the heteroaryl is substituted with one RhC or -(L)bg-RhC (such as RhC or —CH2RhC) and further optionally substituted with from 1-2 RcC; and
    • C6 aryl substituted with one RhC or -(Lg)bg-RhC (such as RhC or —CH2RhC) and further optionally substituted with from 1-2 RcC, wherein each RcC is an independently selected Rc, and each RhC is an independently selected Rh.


In certain of these embodiments, Ring C4 is




embedded image


wherein nc is 0 or 1, such as 0; each RcC is an independently selected Rc, and each RhC is an independently selected Rh.


In certain embodiments, Ring C4 is




embedded image


or wherein Ring C4 is




embedded image


wherein nc is 0 or 1; each RcC is an independently selected Rc, and each RhC is an independently selected Rh.


In certain embodiments, RhC is selected from the group consisting of:

    • C3-8 cycloalkyl which is optionally substituted with from 1-4 Ri; and
    • heterocyclyl of 3-8 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(O)0-2, and wherein the heterocyclyl is optionally substituted with from 1-4 Ri.


In certain embodiments, RhC is




embedded image


wherein XC is N or CH, such as




embedded image


wherein each Ri is an independently selected halo, such as —F.


In some embodiments, Ring C4 is selected from the group consisting of: C3-8 cycloalkyl which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and RcC, and


heterocyclyl of 3-8 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(O)0-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and RcC, wherein each RcC is an independently selected Rc.


In certain embodiments, Ring C4 is C3-8 cycloalkyl which is optionally substituted with from 1-4 RcC, such as C3, C4, C5, or C6 cycloalkyl optionally substituted with from 1-2 RcC, such as unsubstituted C3, C4, C5, or C6 cycloalkyl.


In certain embodiments, each occurrence of RcC is independently selected from the group consisting of: halo; cyano; C1-4 alkyl such as methyl; C1-4 alkyl substituted with from 1-6 independently selected halo, such as —CF3; C1-4 alkoxy, such as methoxy, ethoxy, or isopropoxy; and C1-4 haloalkoxy, such as —OCF3 or —OCHF2.


Non-Limiting Combinations


In certain embodiments, the compound is a compound of Formula (I-a):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L1 is selected from the group consisting of: —O—, —N(H)—, and —N(Rd)—;


L2 is selected from the group consisting of:

    • straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb;
    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


In certain embodiments of Formula (I-a), L1 is —O—.


In certain embodiments of Formula (I-a), L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


In certain embodiments of Formula (I-a), L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—, optionally wherein L2 is —CH2—.


In certain embodiments of Formula (I-a), L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb. In certain of these embodiments, L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -Q1. For example, L2 can be —CH2CH2—.


In certain embodiments of Formula (I-a), L2 is straight-chain C3 alkylene which is optionally substituted with 1-3 Rb.


In certain embodiments of Formula (I-a), L2 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0, 1, or 2; Q2 is CH, CRc, or N; and the asterisk represents the point of attachment to Q1.


In certain of these embodiments, n1 and n2 are independently 0 or 1, optionally 0; and Q2 is CH. For example, n1 and n2 can both be 0; and Q2 can be CH, e.g., L2 can be optionally substituted optionally substituted cyclobutane-diyl, e.g, optionally substituted cyclobutane-1,3-diyl.


In certain embodiments of Formula (I-a), L1 is —O—; and L2 is




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0 or 1, optionally 0; and Q2 is CH. For example, n1 and n2 can both be 0; and Q2 can be CH, e.g., L2 can be optionally substituted cyclobutane-diyl, e.g, optionally substituted 1,3-cyclobutane-1,3-diyl, e.g., unsubstituted cyclobutane-diyl, e.g, unsubstituted cyclobutane-1,3-diyl.


In certain embodiments of Formula (I-a), L1 is —O—; and L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


In certain of the foregoing embodiments of Formula (I-a), L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb.


In certain of the foregoing embodiments, L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -Q1. For example, L2 can be —CH2CH2—.


In certain embodiments of Formula (I-a), L1 is —O—; and L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2. For example, L2 can be —CH2—.


In certain embodiments, the compound is a compound of Formula (I-b):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L2 is straight-chain C1-6 alkylene or straight-chain C2-6 alkenylene, each of which is optionally substituted with 1-6 Rb.


In certain embodiments of Formula (I-b), L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


In certain embodiments of Formula (I-b), L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb. In certain of these embodiments, L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -Q1. For example, L2 can be —CH2CH2—.


In certain embodiments of Formula (I-b), L2 is straight-chain C3 alkylene which is optionally substituted with 1-3 Rb. In certain of these embodiments, L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents point of attachment to -Q1. For example, L2 can be




embedded image


In certain embodiments of Formula (I-b), L2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 Rb.


In certain of these embodiments, L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents the point of attachment to -Q1.


In certain embodiments, the compound is a compound of Formula (I-c):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L2 and L4 are independently selected straight-chain C1-3 alkylene which is optionally substituted with 1-6 Rb; and


L3 is selected from the group consisting of: —O—, —N(H)—, and —N(Rd)—.


In certain embodiments of Formula (I-c), L2 and L4 are independently selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2. In certain of these embodiments, L2 and L4 are each —CH2—.


In certain embodiments of Formula (I-c), L3 is —O—.


In certain embodiments of Formula (I-c), L3 is —N(H)— or —N(Rd)—. For example, L3 can be —N(H)—.


In certain embodiments, the compound is a compound of Formula (I-d):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L2 is straight-chain C1-3 alkylene which is optionally substituted with 1-6 Rb; and


L3 is selected from the group consisting of: —O—, —N(H)—, and —N(Rd)—.


In certain embodiments of Formula (I-d), L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2.


In certain embodiments of Formula (I-d), L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb. In certain of these embodiments, L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -L3. For example, L2 can be —CH2CH2—.


In certain embodiments of Formula (I-d), L3 is —O—.


In certain embodiments of Formula (I-d), L3 is —N(H)— or —N(Rd)—. For example, L3 can be —N(H)—.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is selected from the group consisting of:

    • heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is selected from the group consisting of:

    • heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is phenyl or pyridyl, each optionally substituted with 1-3 Rc′.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is




embedded image


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is phenyl or pyridyl, each optionally substituted with 1-3 Rc′,


wherein each Rc present in Q1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is




embedded image


and each Rc present in Q1 is independently selected from the group consisting of: —F, —Cl, and —CF3.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc′.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is:




embedded image


wherein m1 and m2 are each independently 0, 1, or 2.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is




embedded image


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is:




embedded image


and


the Rd present in Q1 is selected from the group consisting of: —C(O)O(C1-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected Ra; or


wherein the Rd present in Q1 is C2-3 alkyl substituted with 1-3 —F. In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Q1 is




embedded image


and


the Rd present in Q1 is selected from the group consisting of: —C(O)O(C1-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected Ra; or


wherein the Rd present in Q1 is C2-3 alkyl substituted with 1-3 —F.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Rc′ is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 independently selected Rc.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), Rc′ is C6-10 aryl optionally substituted with 1-4 Rc.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), each Ri is H.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), one occurrence of R1 is Rc; and each remaining R1 is H.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), R2 is H; and R1 is H.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W has formula (A-1) as defined in [1] herein and in (i) in the claims and anywhere herein.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W has formula (A-2) as defined in [2] herein and in (ii) in the claims and anywhere herein.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W is heteroaryl of 5 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; provided the heteroaryl is attached to the C(═O)NR6 group via a ring carbon atom as defined in [3] herein and in (iii) in the claims and anywhere herein.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W has formula (A-3) as defined in [4] herein and in (iv) in the claims and anywhere herein.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W has formula (A-3-1) as defined in [4] herein and in (iv) in the claims and anywhere herein.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W bicyclic or polycyclic ring as defined in [5] herein and in (v) in the claims and anywhere herein.


In certain embodiments of Formula (I-a), (I-b), (I-c) or (I-d), W has formula (A-4) as defined in [6] herein and in (vi) in the claims and anywhere herein.


Non-Limiting Exemplary Compounds


In some embodiments, the compound is selected from the group consisting of the compounds delineated in Table C1 or a pharmaceutically acceptable salt thereof.












TABLE C1





Com-





pound





No.
Structure
IUPAC names
LC-MS


















101


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)spiro[2.2]pentane- 1-carboxamide
401.3





102


embedded image


2-cyclopropyl-N- (5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)acetamide
429.3





103


embedded image


1-methyl-N-(5-(2- (4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
429.2





104


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)spiro[2.3]hexane- 1-carboxamide
429.2





105


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)spiro[2.3]hexane- 5-carboxamide
429.2





106


embedded image


3-cyano-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
440.1





107


embedded image


3-methyl-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
429.1





108


embedded image


3-fluoro-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
433.1





109


embedded image


3-fluoro-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)picolinamide
444.0





110


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)thiazole-2- carboxamide
432.0





111


embedded image


Unable to generate value
430.0





112


embedded image


3,5-dimethyl-N-(5- (4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)isoxazole-4- carboxamide
444.1





113


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)thiazole-5- carboxamide
432.0





114


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)thiazole-4- carboxamide
432.0





115


embedded image


Unable to generate value
438.2





116


embedded image


3-fluoro-N-(5-(2- (4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
431.2





117


embedded image


3-fluoro-N-(5-(2- (4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)picolinamide
444.2





118


embedded image


Unable to generate value
430.2





119


embedded image


3,5-dimethyl-N-(5- (2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)isoxazole-4- carboxamide
442.2





120


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)thiazole-5- carboxamide
432.1





121


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)isonicotinamide
426.2





122


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
415.2





123


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)nicotinamide
426.2





124


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)picolinamide
426.2





125


embedded image


2-cyclopropyl-N- (5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)acetamide
403.2





126


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)spiro[2.2]pentane- 1-carboxamide
415.2





127


embedded image


5-methyl-N-(5- ((1r,3r)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1,3,4-thiadiazole- 2-carboxamide
471.1





128


embedded image


1-methyl-N-(5- ((1r,3r)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-1,2,3-triazole- 4-carboxamide
456.1





129


embedded image


3-cyano-N-(5-(((4- (trifluoromethyl)ben- zyl)oxy)methyl)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
457.1





130


embedded image


3-phenyl-N-(5- (((4- (trifluoromethyl)ben- zyl)oxy)methyl)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
489.1





131


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
415.0





132


embedded image


3-fluoro-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
459.1





133


embedded image


3-fluoro-N-(5- ((1r,3r)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
459.1





134


embedded image


3-fluoro-N-(5-(((4- (trifluoromethyl)ben- zyl)oxy)methyl)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
431.15





135


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
413.1





136


embedded image


1-methyl-N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3-yl)-1H- imidazole-2- carboxamide
415.3





137


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
401.2





138


embedded image


3-fluoro-N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
419.2





139


embedded image


1-methyl-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-imidazole-2- carboxamide
455.3





140


embedded image


1-methyl-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-1H- imidazole-2- carboxamide
476.4





141


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)thiazole-2- carboxamide
418.1





142


embedded image


3-methyl-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
476.4





143


embedded image


1-methyl-N-(5-(2- (4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3-yl)-1H- imidazole-2- carboxamide
429.0





144


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)spiro[3.3]heptane- 2-carboxamide
443.3





145


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)oxazole-2- carboxamide
416.2





146


embedded image


1-methyl-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3-yl)-1H- imidazole-2- carboxamide
429.0





147


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)isonicotinamide
412.0





148


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)nicotinamide
412.0





149


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)picolinamide
412.0





150


embedded image


2-cyclopropyl-N- (5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)acetamide
389.3





151


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)spiro[3.3]heptane- 2-carboxamide
429.0





152


embedded image


3-fluoro-N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)picolinamide
430.0





153


embedded image


3,5-dimethyl-N-(5- ((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)isoxazole-4- carboxamide
430.0





154


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)thiazole-5- carboxamide
418.1





155


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)spiro[3.3]heptane- 2-carboxamide
469.0





156


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)spiro[2.3]hexane- 5-carboxamide
455.0





157


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)spiro[3.3]hexane- 2-carboxamide
490.1





158


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)spiro[2.2]pentane- 1-carboxamide
462.1





159


embedded image


5-methyl-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)isoxazole-4- carboxamide
477.4





160


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
415.2





161


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- yl)oxazole-2- carboxamide
416.1





162


embedded image


1-methyl-N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
415.3





163


embedded image


3-methyl-N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
415.3





164


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
401.3





165


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)thiazole-4- carboxamide
418.2





166


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)isonicotinamide
452.3





167


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-pyrazole-5- carboxamide
441.3





168


embedded image


1-methyl-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-pyrazole-5- carboxamide
455.3





169


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)nicotinamide
452.3





170


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)picolinamide
452.3





171


embedded image


3-methyl-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
455.4





172


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)spiro[2.2]pentane- 1-carboxamide
441.3





173


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
441.3





174


embedded image


3-fluoro-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)picolinamide
470.3





175


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)thiazole-2- carboxamide
458.2





176


embedded image


5-methyl-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)isoxazole-4- carboxamide
456.3





177


embedded image


3,5-dimethyl-N-(5- ((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)isoxazole-4- carboxamide
470.3





178


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)thiazole-5- carboxamide
458.3





179


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)isonicotinamide
473.4





180


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
462.4





181


embedded image


1-methyl-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
476.4





182


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)nicotinamide
473.4





183


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)picolinamide
473.4





184


embedded image


2-cyclopropyl-N- (5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl-ethoxy)-1H- indol-3- yl)acetamide
450.4





185


embedded image


3-fluoro-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
480.4





186


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
462.4





187


embedded image


3-fluoro-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)picolinamide
491.4





188


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)thiazole-2- carboxamide
479.3





189


embedded image


3,5-dimethyl-N-(5- (2-((3aR,5r,6aS)- 2-(2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)isoxazole-4- carboxamide
491.4





190


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)thiazole-5- carboxamide
479.3





191


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3- y)spiro[3.3]heptane- 2-carboxamide
443.3





192


embedded image


N-(5-((1s,3s)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)thiazole-4- carboxamide
458.3





193


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)thiazole-4- carboxamide
479.3





194


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)isonicotinamide
426.2





195


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)nicotinamide
426.2





196


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)picolinamide
426.2





197


embedded image


2-cyclopropyl-N- (5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)acetamide
403.2





198


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)spiro[2.3]hexane- 5-carboxamide
429.3





199


embedded image


3-methyl-N-(5-(2- (4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
492.4





200


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)spiro[2.2]pentane- 1-carboxamide
415.2





201


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)thiazole-2- carboxamide
430.1





202


embedded image


N-(5-(2-(4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3- yl)thiazole-4- carboxamide
432.2





203


embedded image


1-methyl-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3-yl)-1H- pyrazole-5- carboxamide
429.2





204


embedded image


3-hydroxy-N-(5- ((cis)-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
457.15





205


embedded image


6-methyl-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-2- oxaspiro[3.3]heptane- 6-carboxamide
506.25





206


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-2- oxaspiro[3.3]heptane- 6-carboxamide
492.25





207


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)-3- oxabicyclo[3.1.0]hex- ane-6- carboxamide
457.25





208


embedded image


6,6-difluoro-N-(5- (2-((3aR,5r,6aS)- 2-(2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)bicyclo[3.1.0]hex- ane-3- carboxamide
512.25





209


embedded image


N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3-yl)-3- oxabicyclo[3.1.0]hex- ane-6- carboxamide
431.05





210


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-3- oxabicyclo[3.1.0]hex- ane-6- carboxamide
478.2





211


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)-2- oxaspiro[3.3]heptane- 6-carboxamide
471.3





212


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
441.3





213


embedded image


4- (trifluoromethyl)- N-(5-(trans-3-(6- (trifluoromethyl)py- ridin-3- yl)cyclobutoxy)- 1H-indol-3- yl)benzamide
NA





214


embedded image


4- (trifluoromethyl)- N-(5-(cis-3-(6- (trifluoromethyl)py- ridin-3- yl)cyclobutoxy)- 1H-indol-3- yl)benzamide
NA





215


embedded image


N-(5-(trans-3-(6- (trifluoromethyl)py- ridin-3- yl)cyclobutoxy)- 1H-indol-3- yl)benzamide
NA





216


embedded image


N-(5-(cis-3-(6- (trifluoromethyl)py- ridin-3- yl)cyclobutoxy)- 1H-indol-3- yl)benzamide
NA





217


embedded image


1-methyl-N-(5- (cis-3-(6- (trifluoromethyl)py- ridin-3- yl)cyclobutoxy)- 1H-indol-3-yl)- 1H-1,2,3-triazole- 4-carboxamide
NA





218


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)benzamide
NA





219


embedded image


1-methyl-N-(5- (trans-3-(6- (trifluoromethyl)py- ridin-3- yl)cyclobutoxy)- 1H-indol-3-yl)- 1H-1,2,3-triazole- 4-carboxamide
NA





220


embedded image


1-methyl-N-(5- (trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-imidazole-2- carboxamide
NA





221


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)spiro[2.3]hexane- 5-carboxamide
415.3





222


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)spiro[2.3]hexane- 5-carboxamide
476.4





223


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)spiro[2.3]hexane- 1-carboxamide
476.4





224


embedded image


N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3- yl)benzamide
NA





225


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)isonicotinamide
NA





226


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-pyrazole-5- carboxamide
NA





227


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)nicotinamide
NA





228


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)picolinamide
NA





229


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)spiro[2.3]hexane- 5-carboxamide
455.3





230


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)spiro[2.3]hexane- 1-carboxamide
455.4





231


embedded image


3-fluoro-N-(5- (trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)picolinamide
NA





232


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)thiazole-2- carboxamide
NA





233


embedded image


5-methyl-N-(5- (trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)isoxazole-4- carboxamide
NA





234


embedded image


3,5-dimethyl-N-(5- (trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)isoxazole-4- carboxamide
NA





235


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)thiazole-5- carboxamide
NA





236


embedded image


N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3- yl)spiro[2.3]hexane- 1-carboxamide
415.2





237


embedded image


1-methyl-N-(5- (trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-pyrazole-5- carboxamide
NA





238


embedded image


3-methyl-N-(5- (trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)bicyclo[1.1.1]pen- tane-1- carboxamide
455.3





239


embedded image


N-(5-(trans-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3- yl)thiazole-4- carboxamide
NA





240


embedded image


1-methyl-N-(5-((4- (trifluoromethyl)ben- zyl)oxy)-1H- indol-3-yl)-1H- 1,2,3-triazole-4- carboxamide
NA





241


embedded image


1-methyl-N-(5- (cis-3-(4- (trifluoromethyl)phe- nyl)cyclobutoxy)- 1H-indol-3-yl)- 1H-1,2,3-triazole- 4-carboxamide
NA





242


embedded image


1-methyl-N-(5-(2- ((3aR,5r,6aS)-2- (2,2,2- trifluoroethyl)octa- hydrocyclopenta[c] pyrrol-5- yl)ethoxy)-1H- indol-3-yl)-1H- 1,2,3-triazole-4- carboxamide
NA





243


embedded image


1-methyl-N-(5-(2- (4- (trifluoromethyl)phe- noxy)ethyl)-1H- indol-3-yl)-1H- 1,2,3-triazole-4- carboxamide
NA





244


embedded image


1-methyl-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3-yl)-1H- 1,2,3-triazole-4- carboxamide
NA





245


embedded image


1-(6-(4,4- difluoropiperidin- 1-yl)-5- fluoropyridin-3- yl)-N-(5-(4- (trifluoromethyl)phe- nethoxy)-1H- indol-3-yl)-1H- 1,2,3-triazole-4- carboxamide
NA





246


embedded image


N-(5-phenoxy-1H- indol-3-yl)-1-(4- (trifluoromethyl)ben- zyl)-1H-pyrazole- 4-carboxamide
NA









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-α-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 α-, β, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-β-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, U K. 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 metabisulfite, 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 al., 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 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0.1 mg/Kg to about 100 mg/Kg; from about 0.1 mg/Kg to about 10 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, 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, weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 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 activity (e.g., e.g., STING 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), 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 neurological disorders 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 telegiectasia; 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; Chiari 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; HTLV-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; Kearns-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; Sjögren'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 vasculopathy with 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 embodiments, 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-dioxygenase (IDO), IL-10, transforming growth factor-β (TGFβ), 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, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD160, HVEM-LIGHT, HVEM-BTLA-CD160, 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 PD1 or PD-L1). 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 topoisomerase. Topoisomerases 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, semisynthetic 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 (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-β), 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-1-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. Pat. No. 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 vasculopathy with 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 biologics (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 biologics (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDIO700, obinutuzumab, vobarilizumab, lulizumab, atacicept, PF-06823859, and lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-α-kinoid, OMS721, RC18, RSLV-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 biologics (e.g., belimumab (Benlysta®), anifrolumab, prezalumab, MEDIO700, vobarilizumab, lulizumab, atacicept, PF-06823859, lupizor, rituximab, BT063, BI655064, BIIB059, aldesleukin (Proleukin®), dapirolizumab, edratide, IFN-α-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, fingolimod, firategrast (SB-683699) (formerly T-0047), GED0301, GLPG0634, GLPG0974, guselkumab, golimumab, GSK1399686, HMPL-004 (Andrographis paniculata extract), IMU-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, farnesoid 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 (B1 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, alpha1-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-β-1a, IFN-β-1b), cladribine, corticosteroids (e.g., methylprednisolone), daclizumab, dimethyl fumarate (Tecfidera®), fingolimod (Gilenya®), fluoxetine, glatiramer acetate (Copaxone®), hydroxychloroquine, ibudilast, idebenone, laquinimod, lipoic acid, losartan, masitinib, MD1003 (biotin), mitoxantrone, montelukast, natalizumab (Tysabri®), NeuroVax™, ocrelizumab, ofatumumab, pioglitazone, and RPC1063.


Non-limiting examples of additional therapeutic agents and/or regimens for treating graft-vs-host disease include abatacept, alemtuzumab, alpha1-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 tocafinib, topical IDP-118, topical M518101, topical calcipotriene and betamethasone dipropionate (e.g., MC2-01 cream and Taclonex®), topical β-3073, topical LEO 90100 (Enstilar®), topical betamethasone dipropriate (Sernivo®), halobetasol propionate (Ultravate®), vitamin D analogues (e.g., calcipotriene (Dovonex®) and calcitriol (Vectical®)), anthralin (e.g., Dritho-scalp® and Dritho-crème®), 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®), Apo805K1, baricitinib, FP187, KD025, prurisol, VTP-43742, XP23829, ZPL-389, CF101 (piclidenoson), LAS41008, VPD-737 (serlopitant), upadacitinib (ABT-494), aprmilast, tofacitibin, cyclosporine (Neoral®, Sandimmune®, Gengraf®), biologics (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, MSB11022, 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®) biologics (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), biologics (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.


on-limiting examples of additional therapeutic agents and/or regimens for treating mucositis include AG013, 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 β-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 ρ-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, 1,8-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, 1H 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

The compounds described herein can be prepared using conventional synthetic methodology known to those of skill in the art.


The following abbreviations have the indicated meanings:

    • Ac=acetyl
    • ADDP=1,1′-(azodicarbonyl)-dipiperidine
    • ACN=acetonitrile
    • Boc20=di-tert-butyl pyrocarbornate
    • Bu=butyl
    • Bz=benzoyl
    • CataCxium A=Bis(adamant-1-yl)(butyl)phosphine
    • CMPB=(Cyanomethylene)tri-n-butylphosphorane
    • DAST=Diethylaminosulphur trifluoride
    • DCE=dichloroethane
    • DCM=dichloromethane
    • DIAD=diisopropyl azodicarboxylate
    • DIEA=N,N-diisopropylethylamine
    • DMA=Dimethylacetamide
    • DMAP=4-dimethylaminopyridine
    • DMF=N,N-dimethylformamide
    • DMF-DMA=N,N-dimethylformamide dimethyl acetal
    • DMSO=dimethyl sulfoxide
    • DPPA=diphenyl azidophosphate
    • Dppf=bis(diphenylphosphino)ferrocene
    • DtBPF=1,1′-Bis[bis(1,1-dimethylethyl)phosphino]ferrocene
    • HATU=2-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium
    • hexafluorophosphate
    • HMDS=1,1,1,3,3,3-Hexamethyldisilazane
    • HPLC=high performance liquid chromatography
    • LAH=Lithium aluminum hydride
    • LC-MS=liquid chromatography—mass spectrometry
    • Me=methyl
    • NMI=1-methylimidazole
    • NMR=nuclear magnetic resonance
    • POT=tris(2-methylphenyl)phosphine
    • Py=pyridine
    • RT=retention time
    • TBS=tert-Butyldimethylsilyl
    • TBUP=Tri-n-butylphosphine
    • TCFH=N,N,N′,N′-tetramethylchloroformamidinium-hexafluorophosphate
    • TEA=trimethylamine
    • TFA=trifluoroacetic acid
    • TFAA=trifluoromethanesulfonic anhydride
    • THF=tetrahydrofuran
    • TMS=Trimethylsilyl
    • T3P=2,4,6-tripropyl-2,4,6-trioxo-1,3,5,2,4,6-trioxatriphosphorinane


Examples
Materials and Methods

The LC-MS of schemes 1-7 and Examples 1-11 were recorded using one of the following methods.


LCMS Method A: Kinetex EVO C18 100A, 30*3 mm, 0.5 μL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.30 min, 95% MPB to 10% in 0.10 min.


LCMS Method B: Xselect CSH C18, 50*3 mm, 1.0 μL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.1% FA and Mobile Phase B (MPB): Acetonitrile/0.1% FA. Elution 5% MPB to 100% in 2.00 min, hold at 100% MPB for 0.70 min, 100% MPB to 5% in 0.05 min, then equilibration to 5% MPB for 0.15 min.


LCMS Method C: XBridge Shield RP18, 50*4.6 mm, 0.5 μL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.04% NH3·H2O and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.79 min, 95% MPB to 10% in 0.06 min, then equilibration to 10% MPB for 0.15 min.


LCMS Method D: kinetex 2.6 μm EVO, 50*3 mm, 0.5 μL injection, 1.2 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.70 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min.


LCMS Method E: HALOC18, 30*3 mm, 0.5 μL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.05% TFA and Mobile Phase B (MPB): Acetonitrile/0.05% TFA. Elution 5% MPB to 100% in 1.20 min, hold at 100% MPB for 0.60 min, 100% MPB to 5% in 0.02 min, then equilibration to 5% MPB for 0.18 min.


LCMS Method F: Shim-pack Scepter C18-120, 33*3 mm, 0.5 μL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 50% MPB to 95% in 2.00 min, hold at 95% MPB for 0.60 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min.


LCMS Method G: Poroshell HPH C18, 50 *3 mm, 0.5 μL injection, 1.2 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3+5 mM NH4OH and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.70 min, 95% MPB to 5% in 0.05 min, then equilibration to 5% MPB for 0.25 min.


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.


The LC-MS of schemes 8-16 and Examples 10-26 were recorded using one of the following methods.


LCMS Method A: Kinetex EVO C18 100A, 30*3 mm, 0.5 μL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.30 min, 95% MPB to 10% in 0.10 min.


LCMS Method B: Xselect CSH C18, 50*3 mm, 1.0 μL injection, 1.2 mL/min flowrate, 90-900 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.1% FA and Mobile Phase B (MPB): Acetonitrile/0.1% FA. Elution 5% MPB to 100% in 2.00 min, hold at 100% MPB for 0.70 min, 100% MPB to 5% in 0.05 min, then equilibration to 5% MPB for 0.15 min.


LCMS Method C: kinetex 2.6 μm EVO, 50*3 mm, 0.5 μL injection, 1.2 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 10% MPB to 95% in 2.00 min, hold at 95% MPB for 0.70 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min.


LCMS Method D: HALOC18, 30*3 mm, 0.5 μL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.05% TFA and Mobile Phase B (MPB): Acetonitrile/0.05% TFA. Elution 5% MPB to 100% in 1.20 min, hold at 100% MPB for 0.60 min, 100% MPB to 5% in 0.02 min, then equilibration to 5% MPB for 0.18 min.


LCMS Method E: Shim-pack Scepter C18-120, 33*3 mm, 0.5 μL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/5 mM NH4HCO3 and Mobile Phase B (MPB): Acetonitrile. Elution 50% MPB to 95% in 2.00 min, hold at 95% MPB for 0.60 min, 95% MPB to 10% in 0.05 min, then equilibration to 10% MPB for 0.25 min.


LCMS Method F: Luna Omega PS C18, 33*3 mm, 0.5 μL injection, 1.5 mL/min flowrate, 30-2000 amu scan range, 254 nm UV detection. Mobile Phase A (MPA): Water/0.1% FA and Mobile Phase B (MPB): Acetonitrile/0.1% FA. Elution 5% MPB to 100% in 1.20 min, hold at 100% MPB for 0.60 min, 100% MPB to 5% in 0.02 min, then equilibration to 5% MPB for 0.25 min.


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.


The abbreviation of chemical terms, LCMS and HPLC conditions of Examples 27-51 are listed below.


Abbreviation of Chemical Terms


ACN=Acetonitrile


DCM=Dichloromethane


DMF=Dimethyl Formamide


HATU=2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethyluronium


hexafluorophosphate(V)


TEA=Triethylamine


TFA=trifluoroacetic acid


H2O=Water


FA=Formic acid


HPLC=high-performance liquid chromatography


LCMS=liquid chromatography—mass spectrometry


NMR=nuclear magnetic resonance


Speedvac=Savant SC250EXP SpeedVac Concentrator


DMSO=Dimethyl Suifoxide


LCMS Analysis Condition

    • Method A
    • Instrument: Agilent LCMS system equipped with DAD and ELSD detector
    • Ion mode: Positive
    • Column: Waters X-Bridge C18, 50*2.1 mm*5 μm or equivalent
    • Mobile Phase: A: H2O (0.04% TFA); B: CH3CN (0.02% TFA)
    • Gradient: 4.5 min gradient method, actual method would depend on clogP of compound.
    • Flow Rate: 0.6 mL/min or 0.8 mL/min
    • Column Temp: 40° C. or 50° C.
    • UV: 220 nm
    • Method B
    • Instrument: Agilent LCMS system equipped with DAD and ELSD detector
    • Ion mode: Positive
    • Column: Waters X-Bridge ShieldRP18, 50*2.1 mm*5 μm or equivalent
    • Mobile Phase:A: H2O (0.05% NH3H2O) or 10 mM ammonia bicarbonate; B: CH3CN
    • Gradient: 4.5 min gradient method; actual method would depend on the clogP of the compound.
    • Flow Rate: 0.6 mL/min or 0.8 mL/min
    • Column Temp: 40° C.
    • UV: 220 nm


Prep. HPLC condition


Instrument:


1. GILSON 281 and Shimadzu LCMS 2010A


2. GILSON 215 and Shimadzu LC-20AP


3. GILSON 215


Mobile phase:


A: NH4OH/H2O=0.05% v/v; B: ACN


A: FA/H2O=0.225% v/v; B: ACN


Column


Xtimate C18 150*25 mm*5 μm


Flow rate: 25 mL/min or 30 mL/min


Monitor wavelength: 220&254 nm


Gradient: actual method would depend on clog P of compound


Detector: MS Trigger or UV


PREPARATIVE EXAMPLES

Scheme for the Preparation of Key Intermediates: Schemes Below Illustrate the Preparation of Key Intermediates.




embedded image


Step 1: tert-butyl N-(5-[2-[1-(2,2,2-trifluoroethyl)piperidin-4-yl]ethoxy]-1H-indol-3-yl)carbamate

tert-Butyl N-(5-hydroxy-1H-indol-3-yl)carbamate (300.0 mg, 1.2 mmol, 1.0 equiv.) was dissolved in DCM (20 mL) and cooled to 0° C., then 2-[1-(2,2,2-trifluoroethyl)piperidin-4-yl]ethanol (306.3 mg, 1.5 mmol, 1.2 equiv.) and P(n-Bu)3 (733.4 mg, 3.6 mmol, 3.0 equiv.) were added under an atmosphere of nitrogen. This was followed by the dropwise addition of a solution of ADDP (609.8 mg, 2.4 mmol, 2.0 equiv.) in DCM (5 mL), maintaining the solution at 0° C. The reaction mixture was stirred for 4 hours at ambient temperature and then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl N-(5-[2-[1-(2,2,2-trifluoroethyl)piperidin-4-yl]ethoxy]-1H-indol-3-yl)carbamate (285.0 mg) as a pale yellow solid. LCMS Method C: [M+H]+=442.


Step 2: 5-(2-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethoxy)-1H-indol-3-amine hydrochloride

tert-Butyl N-(5-[2-[1-(2,2,2-trifluoroethyl)piperidin-4-yl]ethoxy]-1H-indol-3-yl)carbamate (1.0 g, 2.3 mmol, 1.0 equiv.) was dissolved in HCl/1,4-dioxane (4N, 10 mL). The reaction mixture was stirred for 40 min at ambient temperature and then concentrated under vacuum to give 5-(2-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethoxy)-1H-indol-3-amine hydrochloride (910.0 mg) as a yellow solid. LCMS Method A: [M+H]+=342.


The intermediate in the following table was prepared using the same method described for Intermediate 1.















Intermediate
Starting material
Structure
LCMS data







Intermediate 2


embedded image




embedded image


Method C: MS-ESI: 321 [M − H]











embedded image


Step 1: N1-(5-bromo-1H-indol-3-yl)-N2-methyloxalamide

5-Bromo-1H-indol-3-amine (1.7 g, 8.0 mmol, 1.0 equiv.) was dissolved in THF (20 mL), then TEA (3.3 mL, 24.1 mmol, 3.0 equiv.), 2-(methylamino)-2-oxoacetic acid (830.2 mg, 8.0 mmol, 1.0 equiv.) and T3P (50% wt., 3.84 g, 12.0 mmol, 1.5 equiv.) were added. The reaction mixture was stirred for 30 min at ambient temperature, then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:1) to give N1-(5-bromo-1H-indol-3-yl)-N2-methyloxalamide (1.2 g) as a brown solid. LCMS Method A: [M+H]+=296.


Step 2: tert-butyl 5-bromo-3-(2-(methylamino)-2-oxoacetamido)-1H-indole-1-carboxylate

N1-(5-Bromo-1H-indol-3-yl)-N2-methyloxalamide (1.2 g, 4.0 mmol, 1.0 equiv.) was dissolved in DCM (12 mL), then DMAP (50.0 mg, 0.4 mmol, 0.1 equiv.) and (Boc)2O (1.0 g, 4.8 mmol, 1.2 equiv.) were added. The reaction mixture was stirred for 1 hour at ambient temperature, then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:2) to give tert-butyl 5-bromo-3-(2-(methylamino)-2-oxoacetamido)-1H-indole-1-carboxylate (950.0 mg) as a white solid. LCMS Method A: [M+H]+=396


Step 3: tert-butyl 5-(hydroxymethyl)-3-(2-(methylamino)-2-oxoacetamido)-1H-indole-1-carboxylate

tert-Butyl 5-bromo-3-(2-(methylamino)-2-oxoacetamido)-1H-indole-1-carboxylate (900.0 mg, 2.2 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (10 mL), then (tributylstannyl)methanol (1823.2 mg, 5.6 mmol, 2.5 equiv.), butyl di-1-adamanthylphosphine (162.8 mg, 0.4 mmol, 0.20 equiv.) and CataCXium A-Pd-G2 (151.8 mg, 0.2 mmol, 0.1 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was heated to 100° C. for 6 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (2:1) to give tert-butyl 5-(hydroxymethyl)-3-(2-(methylamino)-2-oxoacetamido)-1H-indole-1-carboxylate (750.0 mg) as an off-white solid. LCMS Method C: [M+H]+=348.


The intermediates in the following table were prepared using the same method described for Intermediate 3.















Intermediate
Starting material
Structure
LCMS data







Intermediate 4


embedded image




embedded image


Method C: MS-ESI: 335 [M + H]+











embedded image


Step 1: tert-butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate

tert-Butyl N-(5-bromo-1H-indol-3-yl) carbamate (5.0 g, 16.0 mmol, 1.0 equiv.) was dissolved in DCM (30 mL), then Boc2O (4.2 g, 19.3 mmol, 1.2 equiv.) and DMAP (0.2 g, 1.6 mmol, 0.1 equiv.) were added. The reaction mixture was stirred for 4 hours at ambient temperature, then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate (6.0 g) as a white solid. Method A: [M+H]+=411.


Step 2: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(((4-(trifluoromethyl)benzyl)oxy)methyl)-1H-indole-1-carboxylate

tert-Butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate (4.0 g, 9.7 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (50 mL), then tributyl({[4-(trifluoromethyl)phenyl] methoxy} methyl)stannane (4.6 g, 9.7 mmol, 1.0 equiv.), Pd(PPh3)4 (1.1 g, 1.0 mmol, 0.1 equiv.) and LiCl (0.8 g, 19.4 mmol, 2.0 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was heated to 90° C. overnight, then cooled to ambient temperature. The solids were removed by filtration, and the filtrate was concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:3) to give tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(((4-(trifluoromethyl) benzyl) oxy) methyl)-1H-indole-1-carboxylate (1.0 g) as an off-white solid. LCMS Method A: [M+H]+=521.


Step 3: 5-(((4-(trifluoromethyl)benzyl)oxy)methyl)-1H-indol-3-amine

tert-Butyl 3-((tert-butoxycarbonyl) amino)-5-(((4-(trifluoromethyl) benzyl) oxy) methyl)-1H-indole-1-carboxylate (500.0 mg, 0.9 mmol, 1.0 equiv.) was dissolved in DCM (5 mL), then 2,6-lutidine (308.8 mg, 2.9 mmol, 3.0 equiv.) and TMSOTf (640.4 mg, 2.9 mmol, 3.0 equiv.) were added. The reaction mixture was stirred overnight at ambient temperature, then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with dichloromethane/MeOH (10:1) to give 5-({[4-(trifluoromethyl)phenyl]methoxy}methyl)-1H-indol-3-amine (100.0 mg) as a brown solid. LCMS Method A: [M+H]+=321.




embedded image


Step 1: tert-butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate

tert-Butyl (5-bromo-1H-indol-3-yl)carbamate (5.0 g, 16.1 mmol, 1.0 equiv.) was dissolved in THF (80.0 mL), then (Boc)2O (4.2 g, 19.3 mmol, 1.2 equiv.), DMAP (0.2 g, 1.6 mmol, 0.1 equiv.) and TEA (4.6 mL, 32.1 mmol, 2.0 equiv.) were added. The reaction mixture was stirred for 4 hours at ambient temperature, then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate (6.5 g) as a white solid.


Step 2: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate

tert-Butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate (6.0 g, 14.6 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (100.0 mL), then 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (5.6 g, 21.9 mmol, 1.5 equiv.), Pd(dppf)Cl2 (1.1 g, 1.5 mmol, 0.1 equiv.) and Cs2CO3 (9.5 g, 29.2 mmol, 2.0 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was stirred overnight at 90° C. under nitrogen, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:4) to give tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (6.0 g) as a white solid.


Step 3: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-hydroxy-1H-indole-1-carboxylate

tert-Butyl 3-((tert-butoxycarbonyl)amino)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (6.0 g, 13.1 mmol, 1.0 equiv.) was dissolved in THF (80.0 mL) and cooled to 0° C. Then NaOH (1.6 g, 39.3 mmol, 3.0 equiv.) was added at 0° C., followed by the dropwise addition of H2O2 (30% w.t/wt/, 3.0 g, 26.2 mmol, 2.0 equiv), maintaining the reaction mixture at 0° C. The reaction mixture was stirred for 2 hours at ambient temperature, then quenched by the addition of brine. The resulting resolution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:1) to give tert-butyl 3-((tert-butoxycarbonyl)amino)-5-hydroxy-1H-indole-1-carboxylate (2.2 g) as a grey solid.


Step 4: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate

tert-Butyl 3-((tert-butoxycarbonyl)amino)-5-hydroxy-1H-indole-1-carboxylate (1.0 g, 2.9 mmol, 1.0 equiv.) and cis-3-(4-(trifluoromethyl)phenyl)cyclobutan-1-ol (1.2 g, 5.7 mmol, 2.0 equiv.) were dissolved in THF (20.0 mL) and cooled to 0° C., then TBUP (1.7 g, 8.6 mmol, 3.0 equiv.) was added at 0° C. under an atmosphere of nitrogen. This was followed by the dropwise addition of ADDP (2.2 g, 8.6 mmol, 3.0 equiv.), maintaining the solution at 0° C. The reaction mixture was heated to 50° C. for 2 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase A: 0.05% NH4HCO3 in water; mobile phase B: Acetonitrile, 45% phase B to 70% gradient in 20 min; detector, UV 254 nm. This gave tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate (1.2 g) as an off-white solid.


Step 5: 5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-amine TFA salt

tert-Butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate (190.0 mg, 0.3 mmol, 1.0 equiv.) was dissolved in DCM (2.0 mL), then TFA (2.0 mL) was added. The resulting mixture was stirred for 1 hour at ambient temperature and then concentrated under vacuum to give 5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-amine TFA salt (120.0 mg) as a white solid. LCMS Method A: [M+H]+=347.


The intermediates in the following table were prepared using the same method described for Intermediate 6.















Intermediate
Starting material
Structure
LCMS data







Intermediate 7


embedded image




embedded image


Method A: MS-ESI: 347 [M + H]+











embedded image


Step 3: tert-butyl N-(5-[2-[4-(trifluoromethyl)phenoxy]ethyl]-1H-indol-3-yl)carbamate

tert-Butyl N-[5-(2-hydroxyethyl)-1H-indol-3-yl]carbamate (338.0 mg, 1.2 mmol, 1.0 equiv.) and 4-(trifluoromethyl)phenol (198.2 mg, 1.2 mmol, 1.0 equiv.) were dissolved in THF (10 mL), then ADDP (612.4 mg, 2.4 mmol, 2.0 equiv.) and TBUP (494.9 mg, 2.4 mmol, 2.0 equiv.) were added. The reaction mixture was heated to 70° C. for 5 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:1) to give tert-butyl N-(5-[2-[4-(trifluoromethyl)phenoxy]ethyl]-1H-indol-3-yl)carbamate (260.0 mg) as a brown solid. LCMS Method A: [M+H]+=421.


Step 4: 5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-1H-indol-3-amine TFA salt

tert-Butyl N-(5-{2-[4-(trifluoromethyl)phenoxy]ethyl}-1H-indol-3-yl)carbamate (260.0 mg, 0.6 mmol, 1.0 equiv.) was dissolved in DCM (2 mL) and TFA (2 mL). The reaction mixture was stirred for 30 min at ambient temperature then concentrated under vacuum to give 5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-1H-indol-3-amine TFA salt (350.0 mg) as a yellow solid. LCMS Method A: [M+H]+=321.




embedded image


Step 1: 1-(tert-butyl) 3-methyl 5-bromo-1H-indole-1,3-dicarboxylate

Methyl 5-bromo-1H-indole-3-carboxylate (5.0 g, 19.7 mmol, 1.0 equiv.) and DMAP (0.24 g, 1.9 mmol, 0.1 equiv.) were dissolved in DCM (50 mL), then a solution of (Boc)2O (6.4 g, 29.1 mmol, 1.5 equiv.) in DCM (5 mL) was added dropwise. The reaction mixture was stirred for 4 hours at ambient temperature then quenched by the addition of water. The resulting solution was extracted with dichloromethane, washed with brine, dried over anhydrous Na2SO4 then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:7) to give 1-(tert-butyl) 3-methyl 5-bromo-1H-indole-1,3-dicarboxylate (6.2 g) as a white solid. LCMS Method A: [M+H]+=354.


Step 2: 1-(tert-butyl) 3-methyl 5-vinyl-1H-indole-1,3-dicarboxylate

1-(tert-Butyl) 3-methyl 5-bromo-1H-indole-1,3-dicarboxylate (6.5 g, 18.3 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (60 mL), then tributyl(ethenyl)stannane (13.3 g, 41.8 mmol, 2.3 equiv.), Butyldi-1-adamantylphosphine (1.3 g, 3.6 mmol, 0.2 equiv.) and CataCXium A-Pd-G2 (0.1 g, 0.1 mmol, 0.1 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was heated to 100° C. for 4 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:9) to give 1-(tert-butyl) 3-methyl 5-vinyl-1H-indole-1,3-dicarboxylate (2.8 g) as a yellow solid. LCMS Method A: [M+H]+=302.


Step 3: methyl 5-(3-oxocyclobutyl)-1H-indole-3-carboxylate

DMA (1.1 mL, 11.9 mmol, 1.2 equiv.) was dissolved in DCE (50 mL) and cooled to 5° C., then Tf2O (2.0 mL, 11.9 mmol, 1.2 equiv.) was added dropwise, maintaining the solution at 5° C. The reaction mixture was stirred for 30 min at 5° C. This was followed by the dropwise addition of a solution of 1-(tert-butyl) 3-methyl 5-vinyl-1H-indole-1,3-dicarboxylate (3.0 g, 9.9 mmol, 1.0 equiv.) in DCE (10 mL) at 5° C. The resulting mixture was heated to 80° C. overnight, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:2) to give methyl 5-(3-oxocyclobutyl)-1H-indole-3-carboxylate (300.0 mg) as a yellow solid. LCMS Method A: [M+H]+=244.


Step 4: methyl 5-(cis-3-hydroxycyclobutyl)-1H-indole-3-carboxylate

Methyl 5-(3-oxocyclobutyl)-1H-indole-3-carboxylate (300.0 mg, 1.2 mmol, 1.0 equiv.) was dissolved in MeOH (10 mL) and cooled to 0° C., then NaBH4 (93.3 mg, 2.5 mmol, 2.0 equiv.) was added, maintaining the solution at 0° C. The reaction mixture was stirred for 1 hour at 0° C., then quenched by the addition of water. The resulting solution was extracted with Et2O, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with dichloromethane/MeOH (10:1) to give methyl 5-(cis-3-hydroxycyclobutyl)-1H-indole-3-carboxylate (150.0 mg) as a yellow solid. LCMS Method A: [M+H]+=246.


Step 5: methyl 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carboxylate

Methyl 5-(cis-3-hydroxycyclobutyl)-1H-indole-3-carboxylate (130.0 mg, 0.5 mmol, 1.0 equiv.) and 4-(trifluoromethyl)phenol (129.7 mg, 0.8 mmol, 1.5 equiv.) were dissolved in THF (5 mL) and cooled to 0° C., then TBUP (25.7 mg, 0.1 mmol, 0.2 equiv.) and ADDP (273.3 mg, 1.1 mmol, 2.0 equiv.) were added. The reaction mixture was heated to 70° C. for 4 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:1) to give methyl 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carboxylate (140.0 mg) as a yellow solid. LCMS Method A: [M+H]+=390.


Step 6: 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carboxylic acid

Methyl 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carboxylate (200.0 mg, 0.5 mmol, 1.0 equiv.) was dissolved in MeOH (3 mL), then aqueous NaOH (2 mL, 2M, 4.0 mmol, 8.0 equiv.) was added. The reaction mixture was heated to 70° C. for 2 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was diluted with water and adjusted to pH 4 with aqueous HCl (4M). The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to give 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carboxylic acid (180.0 mg) as a yellow solid. LCMS Method A: [M−H]=374.


Step 7: 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carbonyl azide

5-(trans-3-(4-(Trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carboxylic acid (170.0 mg, 0.4 mmol, 1.0 equiv.) and TEA (0.3 mL, 2.3 mmol, 5.2 equiv.) were dissolved in THF (10 mL), then DPPA (186.9 mg, 0.7 mmol, 1.5 equiv.) was added. The reaction mixture was stirred for 2 hours at ambient temperature, then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:3) to give 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carbonyl azide (150.0 mg) as a yellow solid. LCMS Method A: [M+H]+=401.


Step 8: tert-butyl (5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indol-3-yl)carbamate

5-(trans-3-(4-(Trifluoromethyl)phenoxy)cyclobutyl)-1H-indole-3-carbonyl azide (140.0 mg, 0.3 mmol, 1.0 equiv.) was dissolved in t-BuOH (5 mL). The reaction mixture was heated to 90° C. for 2 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl (5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indol-3-yl)carbamate (100.0 mg) as a yellow solid. LCMS Method A: [M+H]+=447.


Step 9: 5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indol-3-amine TFA salt

tert-Butyl (5-(trans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indol-3-yl)carbamate (90.0 mg, 0.2 mmol, 1.0 equiv.) was dissolved in DCM (5 mL) and TFA (1.5 mL). The reaction mixture was stirred for 2 hours at ambient temperature, then concentrated under vacuum to give 5-(ttrans-3-(4-(trifluoromethyl)phenoxy)cyclobutyl)-1H-indol-3-amine TFA salt (82.0 mg) as a yellow solid. LCMS Method A: [M+H]+=347.




embedded image


Step 1: 3-[4-(trifluoromethyl)phenyl]cyclobutan-1-one

DMA (1.21 g, 13.941 mmol, 1.2 equiv) was dissolved in DCE (30 mL) and cooled to 5° C., then Tf2O (2.7 mL, 16.3 mmol, 1.4 equiv.) was added dropwise, maintaining the solution at 5° C. The reaction mixture was stirred for 30 min at 5° C. This was followed by the addition of a solution of 1-ethenyl-4-(trifluoromethyl) benzene (840.0 mg, 4.9 mmol, 1.0 equiv.) and 2,4,6-collidine (2.0 g, 16.3 mmol, 1.4 equiv.) in DCE (10 mL) was added dropwise at 5° C. The resulting mixture was heated to 80° C. overnight, then cooled to ambient temperature and concentrated under vacuum. The residue was diluted with water, extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. reduced pressure. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:7) to give 3-[4-(trifluoromethyl)phenyl]cyclobutan-1-one (450.0 mg) as a pale yellow oil. 1H NMR (400 MHz, Chloroform-d) δ 7.64 (d, J=8.0 Hz, 2H), 7.45 (d, J=8.0 Hz, 2H), 3.77 (p, J=8.0 Hz, 1H), 3.63-3.50 (m, 2H), 3.34-3.23 (m, 2H).


Step 2: cis-3-[4-(trifluoromethyl)phenyl]cyclobutan-1-ol

3-[4-(Trifluoromethyl)phenyl]cyclobutan-1-one (300.0 mg, 1.4 mmol, 1.0 equiv.) was dissolved in MeOH (15 mL) and cooled to −10° C., then NaBH4 (106.0 mg, 2.8 mmol, 2.0 equiv.) was added, maintaining the solution at −10° C. The reaction mixture was stirred for 50 min at −10° C. under an atmosphere of nitrogen and then quenched by the addition of ice-water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to give cis-3-[4-(trifluoromethyl)phenyl]cyclobutan-1-ol (260.0 mg) as a pale yellow oil. 1H NMR (400 MHz, Chloroform-d) δ 7.58 (d, J=8.0 Hz, 2H), 7.35 (d, J=8.0 Hz, 2H), 4.41-4.29 (m, 1H), 3.10-2.99 (m, 1H), 2.88-2.78 (m, 2H), 2.12-2.00 (m, 2H).


Step 3: trans-3-[4-(trifluoromethyl)phenyl]cyclobutyl 4-nitrobenzoate

Cis-3-[4-(trifluoromethyl)phenyl]cyclobutan-1-ol (130.0 mg, 0.6 mmol, 1.0 equiv.) was dissolved in THF (2 mL), then P-nitrobenzoic acid (100.5 mg, 0.6 mmol, 1.0 equiv.), PPh3 (315.4 mg, 1.2 mmol, 2.0 equiv.) and DIAD (243.2 mg, 1.2 mmol, 2.0 equiv.) were added. The reaction mixture was stirred for 4 hours at ambient temperature and then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:6) to give trans-3-[4-(trifluoromethyl)phenyl]cyclobutyl 4-nitrobenzoate (160.0 mg) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 8.38 (d, J=8.8 Hz, 2H), 8.26 (d, J=8.8 Hz, 2H), 7.71 (d, J=8.0 Hz, 2H), 7.58 (d, J=8.0 Hz, 2H), 5.41 (p, J=6.0 Hz, 1H), 3.88 (p, J=9.2 Hz, 1H), 2.80-2.61 (m, 4H).


Step 4: trans-3-[4-(trifluoromethyl)phenyl]cyclobutan-1-ol

Trans-3-[4-(trifluoromethyl)phenyl]cyclobutyl 4-nitrobenzoate (300.0 mg, 0.8 mmol, 1.0 equiv.) was dissolved in MeOH (4 mL) and water (1 mL), then K2CO3 (227.0 mg, 1.6 mmol, 2.0 equiv.) was added. The reaction mixture was heated to 65° C. for 2 hours, then cooled to ambient temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to give trans-3-[4-(trifluoromethyl)phenyl]cyclobutan-1-ol (155.2 mg) as a pale yellow oil. 1H NMR (400 MHz, DMSO-d6) δ 7.64 (d, J=8.0 Hz, 2H), 7.47 (d, J=8.0 Hz, 2H), 5.14 (d, J=5.6 Hz, 1H), 4.39-4.27 (m, 1H), 3.58 (p, J=7.3 Hz, 1H), 2.39-2.31 (m, 4H), 1.36 (s, OH), 1.23 (s, OH).




embedded image


Step 1: tert-butyl (Z)-5-(2-ethoxy-2-oxoethylidene) hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate

2-(Diethoxyphosphoryl)acetate (28.1 g, 125.2 mmol, 1.2 equiv.) was dissolved in THF (250 mL) and cooled to 0° C., then sodium hydride (6.9 g, 60% wt., 103.5 mmol, 1.0 equiv.) was added in portions under nitrogen atmosphere. After stirred for 15 min, tert-butyl (3aR,6aS)-5-oxohexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate (23.5 g, 104.3 mmol, 1.0 equiv.) was added. The reaction mixture was stirred for 2 hours at room temperature and then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with DCM/petroleum ether (1:1) to give tert-butyl (Z)-5-(2-ethoxy-2-oxoethylidene)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate (16.4 g) as a colorless oil. LCMS Method A: [M+H]+=296.1.


Step 2: tert-butyl trans-5-(2-ethoxy-2-oxoethyl) hexahydrocyclopenta [c]pyrrole-2(1H)-carboxylate

tert-Butyl (Z)-5-(2-ethoxy-2-oxoethylidene)hexahydrocyclopenta [c]pyrrole-2(1H)-carboxylate (16.3 g, 55.3 mmol, 1.0 equiv.) was dissolved in MeOH (200 mL), then Pd/C (10% wt., 2.9 g) was added. The reaction mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred for 4 hours at ambient temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum to give crude tert-butyl trans-5-(2-ethoxy-2-oxoethyl) hexahydrocyclopenta [c]pyrrole-2(1H)-carboxylate (15.5 g) as a colorless oil. LCMS Method A: [M+H]+=298.2.


Step 3: tert-butyl trans-5-(2-hydroxyethyl) hexahydrocyclopenta [c]pyrrole-2 (1H)-carboxylate

tert-Butyl trans-5-(2-ethoxy-2-oxoethyl)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate (13.5 g, 45.4 mmol, 1.0 equiv.) was dissolved in THF (140 mL) and cooled to 0° C., then LiAlH4 (1.7 g, 45.4 mmol, 1.0 equiv.) was added in portions. The reaction mixture was stirred for 1 hour at 0° C. under nitrogen atmosphere and then quenched by the addition of ice-water. The resulting mixture was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to afford tert-butyl trans-5-(2-hydroxyethyl) hexahydrocyclopenta [c]pyrrole-2 (1H)-carboxylate (10.8 g) as a colorless oil. LCMS Method A: [M+H]+=256.2.


Step 4: 2-(trans-octahydrocyclopenta[c]pyrrol-5-yl)ethan-1-ol hydrochloride

tert-Butyl trans-5-(2-hydroxyethyl)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate (10.0 g, 39 mmol, 1.0 equiv.) was dissolved in HCl (gas) in 1,4-dioxane (4M, 50 mL). The reaction mixture was stirred for 1 hour at room temperature and then concentrated under vacuum to afford crude 2-(trans-octahydrocyclopenta [c]pyrrol-5-yl)ethan-1-ol hydrochloride (8.0 g) as a brown solid. LCMS Method A: [M+H]+=156.1.


Step 5: 2-(trans-2-(2,2,2-trifluoroethyl)octahydrocyclopenta [c]pyrrol-5-yl)ethan-1-ol

2-(trans-octahydrocyclopenta [c]pyrrol-5-yl)ethan-1-ol (7.5 g, 48.1 mmol, 1.0 equiv.) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (13.0 g, 57.5 mmol, 1.2 equiv.) were dissolved in ACN (150 mL), then K2CO3 (20.0 g, 144.5 mmol, 3.0 equiv.) was added. The reaction mixture was stirred for 2 hours at 70° C., then cooled to room temperature and quenched by the addition of water. The resulting solution was extracted with ethyl acetate, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with petroleum ether/ethyl acetate (3:1) to give 2-(trans-2-(2,2,2-trifluoroethyl)octahydrocyclopenta [c]pyrrol-5-yl)ethan-1-ol (5.6 g) as a light yellow oil. LCMS Method A: [M+H]+=238.2.




embedded image


Step 1: tert-butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate

tert-Butyl (5-bromo-1H-indol-3-yl)carbamate (5.0 g, 16.1 mmol, 1.0 equiv.) was dissolved in THF (80.0 mL), then (Boc)2O (4.2 g, 19.3 mmol, 1.2 equiv.), DMAP (0.2 g, 1.6 mmol, 0.1 equiv.) and TEA (4.6 mL, 32.1 mmol, 2.0 equiv.) were added. The reaction mixture was stirred for 4 hours at ambient temperature, then concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:5) to give tert-butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate (6.5 g) as a white solid. LCMS Method A: [M+H]+=411.3.


Step 2: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate

tert-Butyl 5-bromo-3-((tert-butoxycarbonyl)amino)-1H-indole-1-carboxylate (6.0 g, 14.6 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (100.0 mL), then 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (5.6 g, 21.9 mmol, 1.5 equiv.), Pd(dppf)Cl2 (1.1 g, 1.5 mmol, 0.1 equiv.) and Cs2CO3 (9.5 g, 29.2 mmol, 2.0 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was stirred overnight at 90° C. under nitrogen, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:4) to give tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (6.0 g) as a white solid. LCMS Method A: [M+H]+=459.3.


Step 3: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-hydroxy-1H-indole-1-carboxylate

tert-Butyl 3-((tert-butoxycarbonyl)amino)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (6.0 g, 13.1 mmol, 1.0 equiv.) was dissolved in THF (80.0 mL) and cooled to 0° C. Then NaOH (1.6 g, 39.3 mmol, 3.0 equiv.) was added at 0° C., followed by the dropwise addition of H2O2 (30% w.t/wt/, 3.0 g, 26.2 mmol, 2.0 equiv), maintaining the reaction mixture at 0° C. The reaction mixture was stirred for 2 hours at ambient temperature, then quenched by the addition of brine. The resulting resolution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:1) to give tert-butyl 3-((tert-butoxycarbonyl)amino)-5-hydroxy-1H-indole-1-carboxylate (2.2 g) as a grey solid. LCMS Method A: [M+H]+=349.2.


Step 4: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate

tert-Butyl 3-((tert-butoxycarbonyl)amino)-5-hydroxy-1H-indole-1-carboxylate (1.0 g, 2.9 mmol, 1.0 equiv.) and cis-3-(4-(trifluoromethyl)phenyl)cyclobutan-1-ol (1.2 g, 5.7 mmol, 2.0 equiv.) were dissolved in THF (20.0 mL) and cooled to 0° C., then TBUP (1.7 g, 8.6 mmol, 3.0 equiv.) was added at 0° C. under an atmosphere of nitrogen. This was followed by the dropwise addition of ADDP (2.2 g, 8.6 mmol, 3.0 equiv.), maintaining the solution at 0° C. The reaction mixture was heated to 50° C. for 2 hours, then cooled to ambient temperature and concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase A: 0.05% NH4HCO3 in water; mobile phase B: Acetonitrile, 45% phase B to 70% gradient in 20 min; detector, UV 254 nm. This gave tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate (1.2 g) as an off-white solid. LCMS Method A: [M+H]+=547.2.


Step 5: 5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-amine TFA salt

tert-Butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate (190.0 mg, 0.3 mmol, 1.0 equiv.) was dissolved in DCM (2.0 mL), then TFA (2.0 mL) was added. The resulting mixture was stirred for 1 hour at ambient temperature and then concentrated under vacuum to give 5-(trans-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-amine TFA salt (120.0 mg) as a white solid. LCMS Method A: [M+H]+=347.2.


The Intermediates in the Following Table were Prepared Using the Same Method Described for Intermediates 14.















Inter-





mediate
Starting material
Structure
LCMS data







Inter- mediate 15


embedded image




embedded image


Method A: MS-ESI: 347.2 [M + H]+





Inter- mediate 16


embedded image




embedded image


Method B: MS-ESI: 368.2 [M + H]+





Inter- mediate 17


embedded image




embedded image


Method B: MS-ESI: 307.1 [M + H]+





Inter- mediate 18


embedded image




embedded image


Method A: MS-ESI: 321.1 [M + H]+











embedded image


embedded image


Step 1: 3-(benzyloxy)-1-(6-(trifluoromethyl)pyridin-3-yl)cyclobutan-1-ol

5-Bromo-2-(trifluoromethyl)pyridine (4.0 g, 17.6 mmol, 1.0 equiv.) was dissolved in THF (40 mL) and cooled to −70° C., then n-BuLi (2.5M in hexane, 8.5 mL, 21.3 mmol, 1.2 equiv.) added dropwise, maintaining the solution at −70° C. under an atmosphere of nitrogen. After stirred for 30 min at −70° C., 3-(benzyloxy)cyclobutan-1-one (3.7 g, 21.2 mmol, 1.2 equiv.) was added dropwise. The reaction mixture was stirred for additional 2 hours at rt and then quenched by the addition of saturated aqueous NH4Cl. The resulting solution was extracted with EtOAc, washed with brine, dried over anhyd. Na2SO4 and concentrated under vacuum. The residue was purified by reverse flash column with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.5% NH4HCO3), 10% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in 3-(benzyloxy)-1-(6-(trifluoromethyl)pyridin-3-yl)cyclobutan-1-ol (2.7 g) as a pale yellow solid. LCMS Method A: [M+H]+=324.2.


Step 2: 5-(3-(benzyloxy)-1-fluorocyclobutyl)-2-(trifluoromethyl)pyridine

3-(Benzyloxy)-1-(6-(trifluoromethyl)pyridin-3-yl)cyclobutan-1-ol (2.7 g, 8.3 mmol, 1.0 equiv.) was dissolved in DCM (10 mL) and cooled to −70° C., then DAST (2.6 g, 16.6 mmol, 2.0 equiv.) was added dropwise, maintaining the solution at −70° C. under an atmosphere of nitrogen. The reaction mixture was stirred for 2 hours at rt and then quenched by the addition of water. The resulting solution was extracted with EtOAc, washed with brine, dried over anhyd. Na2SO4 and concentrated under vacuum. The residue was purified by reverse flash column with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% NH4HCO3), 10% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in 5-(3-(benzyloxy)-1-fluorocyclobutyl)-2-(trifluoromethyl)pyridine (2.5 g) as a pale yellow solid. LCMS Method A: [M+H]+=326.0.


Step 3: 3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutan-1-ol

5-[3-(Benzyloxy)-1-fluorocyclobutyl]-2-(trifluoromethyl)pyridine (2.0 g, 6.1 mmol, 1.0 equiv.) was dissolved in MeOH (40 ml), then HCOOH (282.9 mg, 6.1 mmol, 1.0 equiv.) was added. This was followed by the addition of Pd/C (10% wt., 130.8 mg) under an atmosphere of nitrogen. The mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred for 4 hours at 40° C. The solids were removed by filtration and the filter cake was washed with MeOH. The combined filtrate was concentrated under vacuum. The residue was purified by reverse flash column with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% NH4HCO3), 10% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in 3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutan-1-ol (1.0 g) as a pale yellow oil. LCMS Method A: [M+H]+=261.0.


Step 4-5: tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indole-1-carboxylate and tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(cis-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indole-1-carboxylate

3-[6-(Trifluoromethyl)pyridin-3-yl]cyclobutan-1-ol (1.0 g, 4.6 mmol, 1.0 equiv.) was dissolved in THF (13 mL), then tert-butyl 3-[(tert-butoxycarbonyl)amino]-5-hydroxyindole-1-carboxylate (1.6 g, 4.6 mmol, 1.0 equiv.), TBUP (1.8 g, 9.2 mmol, 2.0 equiv.) and ADDP (2.3 g, 9.2 mmol, 2.0 equiv.) were added under an atmosphere of nitrogen. The reaction mixture was stirred for 5 hours at 70° C., then cooled to rt and quenched by the addition of water. The resulting solution was extracted with EtOAc, washed with brine, dried over anhyd. Na2SO4 and concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% NH4HCO3), 10% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indole-1-carboxylate (1.0 g) as a pale yellow solid. The mixture was separated by Chiral-HPLC with the following conditions: Column: JW-CHIRAL-Amylose-SA, 20*250 mm, Sum; Mobile Phase A: IPA-HPLC, Mobile Phase B: Hex (0.5% 2M NH3-MeOH)-HPLC; Flow rate: 20 mL/min; Gradient: 90% B to 90% B in 14 min; Wave Length: 220/254 nm; RT1: 8.2 min; RT2: 10.22 min. This resulted in tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(cis-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indole-1-carboxylate (710.0 mg) as a pale yellow solid. LCMS Method B: [M−H]=548. And tert-butyl 3-((tert-butoxycarbonyl)amino)-5-(trans-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indole-1-carboxylate (170.0 mg) as a pale yellow solid. LCMS Method B: [M−H]-=548.1.


Step 5: 5-(trans-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indol-3-amine TFA salt

tert-Butyl 3-[(tert-butoxycarbonyl)amino]-5-[trans-3-[6-(trifluoromethyl)pyridin-3-yl]cyclobutoxy]indole-1-carboxylate (160.0 mg, 0.2 mmol, 1.0 equiv.) was dissolved in DCM (2 mL), then TFA (2 mL) was added. The reaction mixture was stirred for 1 hours at rt and then concentrated under vacuum to give crude 5-(trans-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indol-3-amine TFA salt (103.0 mg) as a red solid. LCMS Method B: [M+H]+=348.2.




embedded image


tert-Butyl 3-[(tert-butoxycarbonyl)amino]-5-[cis-3-[6-(trifluoromethyl)pyridin-3-yl]cyclobutoxy]indole-1-carboxylate (500.0 mg, 0.9 mmol, 1.0 equiv.) was dissolved in DCM (3 mL), then TFA (3 mL) was added. The reaction mixture was stirred for 1 hour at rt and then concentrated under vacuum to give crude 5-(trans-3-(6-(trifluoromethyl)pyridin-3-yl)cyclobutoxy)-1H-indol-3-amine TFA salt (400.0 mg) as a brown solid. LCMS Method B: [M+H]+=348.2.




embedded image


Step 1: 2-methyl-1-nitro-4-phenoxybenzene

4-Fluoro-2-methyl-1-nitrobenzene (10.0 g, 64.4 mmol, 1.0 equiv.) was dissolved in DMF (15 mL), then phenol (9.1 g, 96.6 mmol, 1.5 equiv.) and K2CO3 (22.2 g, 161.1 mmol, 2.50 equiv.) were added. The reaction mixture was stirred at 80° C. for 18 hours, then cooled to room temperature and quenched by the addition of water. The resulting solution was extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with EtOAc/petroleum ether (1:3) to give 2-methyl-1-nitro-4-phenoxybenzene (12.9 g) as a brown solid. GCMS=229.


Step 2: (E)-N, N-dimethyl-2-(2-nitro-5-phenoxyphenyl)ethen-1-amine

2-Methyl-1-nitro-4-phenoxybenzene (10.0 g, 43.6 mmol, 1.0 equiv.) was dissolved in DMF (20 mL), then DMF-DMA (6.2 g, 52.3 mmol, 1.2 equiv.) was added. The reaction mixture was stirred for 10 hours at 140° C., then cooled to room temperature and concentrated under vacuum. The residue was diluted with water, then extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to give crude (E)-N, N-dimethyl-2-(2-nitro-5-phenoxyphenyl)ethen-1-amine (15.0 g) as a red solid. LCMS Method A: [M+H]+=285.1.


Step 3: 5-phenoxy-1H-indole

(E)-N, N-dimethyl-2-(2-nitro-5-phenoxyphenyl)ethen-1-amine (15.0 g, 52.7 mmol, 1.0 equiv.) was dissolved in EtOAc (20 mL), then Pd/C (2.9 g, 27.9 mmol, 0.5 equiv.) was added. The mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred overnight at room temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:3) to give 5-phenoxy-1H-indole (4 g) as a brown solid. LCMS Method A: [M+H]+=210.1.


Step 4: 3-nitro-5-phenoxy-1H-indole

AgNO3 (4.8 g, 28.6 mmol, 1.5 equiv.) was dissolved in ACN (8 mL) and cooled to 0° C., then benzoyl chloride (4.0 g, 28.6 mmol, 1.5 equiv.) was added at 0° C. The reaction mixture was stirred for 30 minutes at 0° C. To the above mixture was added 5-phenoxy-1H-indole (4.0 g, 19.1 mmol, 1.0 equiv.) and the resulting mixture was stirred for additional 1 hour at room temperature. The reaction was quenched by the addition of ice-water. The resulting solution was extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with EtOAc/petroleum ether (1:3) to give 3-nitro-5-phenoxy-1H-indole (2.5 g) as a brown solid. LCMS Method A: [M−H]-=253.1.


Step 5: tert-butyl (5-phenoxy-1H-indol-3-yl)carbamate

3-Nitro-5-phenoxy-1H-indole (3.0 g, 11.8 mmol, 1.0 equiv.) was dissolved in MeOH (5 mL), then (Boc)2O (3.8 g, 17.7 mmol, 1.5 equiv.) and Pd/C (600.0 mg, 5.6 mmol, 0.4 equiv.) were added. The mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred overnight at room temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:4) to give tert-butyl (5-phenoxy-1H-indol-3-yl)carbamate (1.5 g) as a light pink solid. LCMS Method A: [M+H]+=325.2.


Step 6: 5-phenoxy-1H-indol-3-amine hydrochloride

tert-Butyl (5-phenoxy-1H-indol-3-yl)carbamate (1.5 g, 4.6 mmol, 1.0 equiv.) was dissolved in HCl/1,4-dioxane (4N, 5 mL). The resulting solution was stirred for 30 min at room temperature and concentrated under vacuum to give 5-phenoxy-1H-indol-3-amine hydrochloride (1.0 g) as a brown solid. LCMS Method A: [M+H]+=225.1.




embedded image


Step 1: ethyl 6-methyl-2-oxaspiro[3.3]heptane-6-carboxylate

Ethyl 2-oxaspiro[3.3]heptane-6-carboxylate (300.0 mg, 1.7 mmol, 1.0 equiv.) was dissolved in THF (10 mL) and cooled to −78° C., then LDA (2M in THF, 1.3 mL, 2.6 mmol, 1.5 egyiv.) was added dropwise with stirring at −78° C. under nitrogen atmosphere. The reaction mixture was stirred for 1 hour at −78° C. under nitrogen atmosphere. To the above mixture was added methyl iodide (750.5 mg, 5.3 mmol, 3.0 equiv.) and the resulting mixture was stirred for additional 4 hours at room temperature, then quenched by the addition of saturated aqueous NH4Cl. The resulting solution was extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. This resulted in crude ethyl 6-methyl-2-oxaspiro[3.3]heptane-6-carboxylate (360 mg) as a yellow crude oil.


Step 2: 6-methyl-2-oxaspiro[3.3]heptane-6-carboxylic acid

Ethyl 6-methyl-2-oxaspiro[3.3]heptane-6-carboxylate (360.0 mg, 2.0 mmol, 1.0 equiv.) was dissolved in MeOH (5 mL), then a solution of NaOH in water (2M, 3 mL) was added. The reaction mixture was stirred for 1 hour at 70° C., then cooled to room temperature and concentrated under vacuum. The residue was diluted with water (20 mL), adjusted to pH 5 with HCl aqueous. The resulting solution was extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. This resulted in 6-methyl-2-oxaspiro[3.3]heptane-6-carboxylic acid (240 mg, 78.6%) as a yellow oil. LCMS Method A: [M−H]=155.1.




embedded image


Step 1: ethyl 1-[[4-(trifluoromethyl)phenyl]methyl]pyrazole-4-carboxylate

Ethyl 1H-pyrazole-4-carboxylate (300.0 mg, 2.1 mmol, 1.0 equiv.) and 1-(bromomethyl)-4-(trifluoromethyl)benzene (511.7 mg, 2.1 mmol, 1.0 equiv.) were dissolved in DMF (6 mL), then Cs2CO3 (2.1 g, 6.4 mmol, 3.0 equiv.) was added. The reaction mixture was stirred overnight at ambient temperature and then quenched by the addition of water. The resulting solution was extracted with ethyl acetate and concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, C18 silica gel; mobile phase, water (NH4HCO3. 1 g/l L) and ACN, 0% ACN increasing to 100% ACN within 25 min; Detector, UV 254 nm. This gave ethyl 1-[[4-(trifluoromethyl)phenyl]methyl]pyrazole-4-carboxylate (310 mg) as a white solid. LCMS Method A: [M+H]+=299.1.


Step 2: 1-[[4-(trifluoromethyl)phenyl]methyl]pyrazole-4-carboxylic acid

Ethyl 1-[[4-(trifluoromethyl)phenyl]methyl]pyrazole-4-carboxylate (200.0 mg, 0.7 mmol, 1.0 equiv.) was dissolved in MeOH (2 mL) and water (2 mL), then NaOH (53.6 mg, 1.3 mmol, 2.0 equiv.) was added. The reaction mixture was stirred for 1 hour at ambient temperature and then concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, C18 silica gel; mobile phase, water (10 mM NH4HCO3) and ACN, 0% ACN increasing to 100% within 25 min; Detector, UV 254 nm. This gave 1-[[4-(trifluoromethyl)phenyl]methyl]pyrazole-4-carboxylic acid (130 mg) as a white solid. LCMS Method B: [M−H]=269.1.




embedded image


Step 1: 2-(4,4-difluoropiperidin-1-yl)-3-fluoro-5-nitropyridine

2-Chloro-3-fluoro-5-nitropyridine (10.0 g, 56.6 mmol, 1.0 equiv.) was dissolved in DMF (150 mL), then Cs2CO3 (37.3 g, 114.5 mmol, 2.0 equiv.) and 4,4-difluoropiperidine (9.8 g, 81.0 mmol, 1.4 equiv.) were added. The reaction mixture was heated to 90° C. for hours, then cooled to ambient temperature and quenched by the addition of water. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:3) to give 2-(4,4-difluoropiperidin-1-yl)-3-fluoro-5-nitropyridine (13.3 g) as a yellow solid. LCMS Method D: [M+H]+=262.


Step 2: 6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-amine

2-(4,4-difluoropiperidin-1-yl)-3-fluoro-5-nitropyridine (13.2 g, 50.5 mmol, 1.0 equiv.) was dissolved in MeOH (100 mL), then Pd/C (10% wt., 2.0 g) was added. The mixture was sparged with nitrogen, placed under an atmosphere of hydrogen gas (balloon), then stirred for 15 hours at ambient temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with dichloromethane/methanol (97:3) to give 6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-amine (11.4 g) as a yellow solid. LCMS Method D: [M+H]+=232.


Step 3: 5-azido-2-(4,4-difluoropiperidin-1-yl)-3-fluoropyridine

6-(4,4-Difluoropiperidin-1-yl)-5-fluoropyridin-3-amine (400.0 mg, 1.7 mmol, 1.0 equiv.) was dissolved in ACN (10 mL) and cooled to 0° C., then t-BuNO2 (0.3 mL, 2.7 mmol, 1.6 equiv.) was added dropwise, maintaining the solution at 0° C. The reaction mixture was stirred for 30 min at 0° C. This was followed by the addition of TMSN3 (0.3 mL, 2.5 mmol, 1.5 equiv.) dropwise at 0° C. The resulting mixture was stirred for additional 2 hours at ambient temperature and then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:2) to give 5-azido-2-(4,4-difluoropiperidin-1-yl)-3-fluoropyridine (380.0 mg) as a yellow oil. LCMS Method A: [M+H]+=258.


Step 4: methyl 1-[6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-yl]-1,2,3-triazole-4-carboxylate

5-Azido-2-(4,4-difluoropiperidin-1-yl)-3-fluoropyridine (350.0 mg, 1.4 mmol, 1.0 equiv.) was dissolved in 1,4-dioxane (3.6 mL) and water (0.4 mL), then methyl propiolate (228.8 mg, 2.7 mmol, 2.0 equiv.), sodium (R)-2-((S)-1,2-dihydroxyethyl)-4-hydroxy-5-oxo-2,5-dihydrofuran-3-olate (53.9 mg, 0.3 mmol, 0.2 equiv.) and CuSO4 (21.7 mg, 0.1 mmol, 0.1 equiv.) were added. The reaction mixture was stirred overnight at ambient temperature and then quenched by the addition of water. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1:2) to give methyl 1-[6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-yl]-1,2,3-triazole-4-carboxylate (150.0 mg) as a yellow solid. LCMS Method A: [M+H]+=341.


Step 5: 1-[6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-yl]-1,2,3-triazole-4-carboxylic acid

Methyl 1-[6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-yl]-1,2,3-triazole-4-carboxylate (300.0 mg, 0.9 mmol, 1.0 equiv.) was dissolved in MeOH (3 mL) and water (7 mL), then NaOH (70.3 mg, 1.8 mmol, 2.0 equiv.) was added. The reaction mixture was heated to 80° C. for 2 hours, and then cooled to ambient temperature and concentrated under vacuum. The residue was diluted with water, adjusted to pH 6 with 1M aqueous HCl. The resulting solution was extracted with ethyl acetate, washed with brine, dried over anhydrous Na2SO4 and concentrated under vacuum to afford 1-[6-(4,4-difluoropiperidin-1-yl)-5-fluoropyridin-3-yl]-1,2,3-triazole-4-carboxylic acid (200.1 mg) as a yellow solid. LCMS Method A: [M+H]+=328.


Example 1: trans-N-(5-(-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)bicyclo[1.1.1]pentane-1-carboxamide (Compound 135)



embedded image


5-(trans-3-(4-(Trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-amine TFA salt is dissolved in THF, then bicyclo[1.1.1]pentane-1-carboxylic acid, HATU and DIEA are added. The reaction mixture is stirred for 1 hour at ambient temperature and then concentrated under vacuum. The residue is purified by reverse flash chromatography. This gives trans-N-(5-(-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)bicyclo[1.1.1]pentane-1-carboxamide as the product. The analogs prepared in the following table were prepared using the same method described for Example 1.


















Starting




Example
Compound
materials




#
No.
Used
Structure
LCMS data







1
135
Intermediate 8/bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method G: MS-ESI: 415 [M + H]+.





2
133
Intermediate 6/3-fluoro- bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method E: MS-ESI: 459 [M + H]+.





3
132
Intermediate 7/3-fluoro- bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method E: MS-ESI: 459 [M + H]+.





4
130
Intermediate 5/3-phenyl- bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method D: MS-ESI: 489 [M − H].





5
129
Intermediate 5/3-cyano- bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method G: MS-ESI: 440 [M + H]+.





6
131
Intermediate 2/bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method G: MS-ESI: 415 [M + H]+.





7
134
Intermediate 5/3-fluoro- bicyclo- [1.1.1]- pentane-1- carboxylic acid


embedded image


Method F: MS-ESI: 431 [M − H].





8
128
Intermediate 6/1-methyl- 1H-1,2,3- triazole-4- carboxylic acid


embedded image


Method D: MS-ESI: 456 [M + H]+.





9
127
Intermediate 6/lithium 5- methyl-1,3,4- thiadiazole- 2-carboxylate


embedded image


Method F: MS-ESI: 473 [M + H]+.









Example 10: 3-hydroxy-N-(5-(cis-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3 yl)bicyclo[1.1.1]pentane-1-carboxamide (Compound 204)



embedded image


5-(cis-3-(4-(Trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-amine TFA salt (385.0 mg, 0.8 mmol, 1.0 equiv.) and 3-hydroxybicyclo[1.1.1]pentane-1-carboxylic acid (110.9 mg, 0.8 mmol, 1.0 equiv.) were dissolved in DCM (5 mL), then HATU (494.0 mg, 1.3 mmol, 1.5 equiv.) and DIEA (335.8 mg, 2.6 mmol, 3.0 equiv.) were added. The reaction mixture was stirred for 1 hour at room temperature and then quenched by the addition of water. The resulting solution was extracted with DCM, washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HHPLC with the following conditions: Column: SunFire Prep C18 OBD Column, 19*150 mm, mm; Mobile Phase A: Water (10 mmol/L NH4HCU3), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 60% B to 80% B in 5.3 mMS; Wave Length: 210/254 nm; RT1: 5.3 min. This resulted in 3-hydroxy-N-(5-(cis-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)bicyclo[1.1.1]pentane-1l-carboxamide (144.7 mg, 36.6%) as an off-white solid. LCMS Method F: [M+H]+=457.2. 1H NMR (400 MHz, DMSO-d6) δ 10.68 (s, 1H), 9.24 (s, 1H), 7.69 (d, J=8.0 Hz, 2H), 7.56-7.52 (m, 3H), 7.24-7.19 (m, 2H), 6.76-6.73 (m, 1H), 6.35 (s, 1H), 4.73-4.69 (m, 1H), 3.33-3.29 (m, 1H), 3.00-2.93 (m, 2H), 2.20-2.18 (m, 2H), 2.16-2.13 (in, 6H).


The Analogs Prepared in Table 2 were Prepared Using the Same Method Described for Example 10.













TABLE 2






Starting






materials





Compound
Used
Structure
Condition
LCMS data







Example 11/ Compound 205
Intermediate 16/ Intermediate 23


embedded image


HATU, DIEA, DCM
Method F: MS-ESI: 506.3 [M + H]+.





Example 12/ compound 206
Intermediate 16/Sodium 2-oxaspiro- [3.3]heptane- 6-carboxylate


embedded image


TCFH, NMI, ACN
Method E: MS-ESI: 492.3 [M + H]+.





Example 13/ compound 208
Intermediate 16/6,6- Difluoro- bicyclo- [3.1.0]- hexane-3- carboxylic acid


embedded image


HATU, DIEA, DCM
Method F: MS-ESI: 512.3 [M + H]+.





Example 14/1 compound 207
Intermediate 14/3- oxabicyclo- [3.1.0] hexane-6- carboxylic acid


embedded image


TCFH, NMI, ACN
Method F: MS-ESI: 457.3 [M + H]+.





Example 15/ compound 209
Intermediate 18/3- oxabicyclo- [3.1.0] hexane-6- carboxylic acid


embedded image


HATU, DIEA, DCM
Method E: MS-ESI: 431.1 [M + H]+.





Example 16/ compound 210
Intermediate 16/3- oxabicyclo- [3.1.0] hexane-6- carboxylic acid


embedded image


HATU, DIEA, DCM
Method E: MS-ESI: 478.2 [M + H]+.





Example 17/ compound 246
Intermediate 22/ Intermediate 24


embedded image


HATU, DIEA, DMF
Method D: MS-ESI: 477.1 [M + H]+.





Example 18/ compound 245
Intermediate 18/ Intermediate 25


embedded image


PyBOP, NMM, DMF
Method D: MS-ESI: 630.2 [M + H]+.





Example 19/ compound 219
Intermediate 19/1- methyl-1H- 1,2,3- triazole-4- carboxylic acid


embedded image


T3P, TEA, THF
Method D: MS-ESI: 457.3 [M + H]+.





Example 20/ compound 217
Intermediate 20/1- methyl-1H- 1,2,3- triazole-4- carboxylic acid


embedded image


T3P, TEA, THF
Method D: MS-ESI: 457.3 [M + H]+.









Example 21: N-(5-((4-(trifluoromethyl)benzyl)oxy)-1H-indol-3-yl)benzamide (Compound 218)



embedded image


5-((4-(Trifluoromethyl)benzyl)oxy)-1H-indol-3-amine (350 mg, 1.14 mmol, 1.0 equiv.) and TEA (462.5 mg, 4.6 mmol, 4.0 equiv.) were dissolved in DCM (7 mL), then benzoyl chloride (160.6 mg, 1.1 mmol, 1.0 equiv.) was added at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for 1 hour at room temperature under nitrogen atmosphere and then quenched by the addition of water. The resulting mixture was extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-TLC (petroleum ether/EtOAc=1:1) to afford the crude product, that was further purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 30*150 mm, 5 m; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 49% B to 68% B in 8 min; Wave Length: 254 nm; RT1: 7.3 min. This resulted in N-(5-((4-(trifluoromethyl)benzyl)oxy)-1H-indol-3-yl)benzamide (177.0 mg, 37.7%) as a white solid. LCMS Method E: [M−H]=409.1. 1H NMR (400 MHz, DMSO-d6): δ 10.80 (s, 1H), 10.09 (s, 1H), 8.01-7.98 (m, 2H), 7.81-7.77 (m, 3H), 7.73-7.71 (m, 2H), 7.61-7.52 (m, 4H), 7.29 (d, J=8.8 Hz, 1H), 6.89-6.86 (m, 1H), 5.21 (s, 2H).


The Analogs Prepared in the Following Table were Prepared Using the Same Method Described for Example 21.
















Starting





materials




Compound
Used
Structure
LCMS data







Example 22/ compound 224
Intermediate 16/benzoyl chloride


embedded image


Method C: MS-ESI: 472.1 [M + H]+.





Example 23/ compound 216
Intermediate 20/benzoyl chloride


embedded image


Method C: MS-ESI: 452.1 [M + H]+.





Example 24/ compound 214
Intermediate 20/4- (trifluoro- methyl)- benzoyl chloride


embedded image


Method C: MS-ESI: 520.1 [M + H]+.





Example 25/ compound 215
Intermediate 19/benzoyl chloride


embedded image


Method C: MS-ESI: 452.1 [M + H]+.





Example 26/ compound 213
Intermediate 19/4- (trifluoro- methyl)- benzoyl chloride


embedded image


Method C: MS-ESI: 520.1 [M + H]+.









Example 27. Synthesis of 1-methyl-N-(5-(4-(trifluoromethyl)phenethoxy)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 244)



embedded image


tert-butyl 3-{[(tert-butoxy)carbonyl]amino}-5-{2-[4-(trifluoromethyl)phenyl]ethoxy}-1H-indole-1-carboxylate (83.2 mg, 0.16 mmol, 1.0 equiv.) was dissolved in DCM (2 mL), and TFA (500 μl) was added in the mixture. The mixture was heated at 30° C. for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and 1-methyl-1H-1,2,3-triazole-4-carboxylic acid (40.64 mg, 0.32 mmol, 2.0 equiv.) were dissolved in DMF (2 mL), then TEA (116 μl, 0.8 mmol, 5.0 equiv.) and HATU (63.84 mg, 0.168 mmol, 1.05 equiv.) were added. The mixture was heated at 30° C. for 16 hours. The crude product was purified by prep HPLC to give 1-methyl-N-(5-(4-(trifluoromethyl)phenethoxy)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (41.72 mg, 0.097 mmol) as a powder. MS-ESI, 430.2 [M+H+]. 1H NMR (400 MHz, DMSO-d6) δ ppm 10.81-10.72 (m, 1H), 10.19 (s, 1H), 8.62 (s, 1H), 7.75-7.70 (m, 1H), 7.71-7.66 (m, 2H), 7.59 (d, 2H), 7.45 (d, 1H), 7.23 (d, 1H), 6.72 (dd, 1H), 4.22 (t, 2H), 4.13 (s, 3H), 3.17 (br t, 2H).


Example 28: Synthesis of 1-methyl-N-(5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 243)



embedded image


tert-butyl (5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-1H-indol-3-yl)carbamate (83.2 mg, 0.16 mmol, 1.0 equiv.) were dissolved in DCM (2 mL), and TFA (500 μl) were added in the mixture. The mixture was heated at 30° C. for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and 1-methyl-1H-1,2,3-triazole-4-carboxylic acid (40.64 mg, 0.32 mmol, 2.0 equiv.) were dissolved in DMF (2 mL), then TEA (116 μl, 0.8 mmol, 5.0 equiv.) and HATU (63.84 mg, 0.168 mmol, 1.05 equiv.) were added. The mixture was heated at 30° C. for 16 hours. The crude product was purified by prep HPLC to give 1-methyl-N-(5-(2-(4-(trifluoromethyl)phenoxy)ethyl)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (17.82 mg, 0.041 mmol) as a powder. MS-ESI, 430.3 [M+H+].


Example 29: Synthesis of N-(5-(2-((3aR,5R,6aS)-2-(2,2,2-trifluoroethyl)octahydrocyclopenta [c]pyrrol-5-yl)ethoxy)-1H-indol-3-yl)spiro[2.3]hexane-1-carboxamide (Compound223)



embedded image


tert-butyl 5-{2-[(3aR,5R,6aS)-2-(2,2,2-trifluoroethyl)-octahydrocyclopenta [c]pyrrol-5-yl]ethoxy}-3-{[(tert-butoxy)carbonyl]amino}-1H-indole-1-carboxylate (96.4 mg, 0.17 mmol, 1.0 equiv.) was dissolved in DCM (3 mL), then TFA (1 mL) was added to the solution. The mixture was heated at 30° C. for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and spiro[2.3]hexane-1-carboxylic acid (42.84 mg, 0.34 mmol, 2.0 equiv.) were dissolved in DMF (2 mL), then TEA (123 μl, 0.85 mmol, 5.0 equiv.) and HATU (68.4 mg, 0.18 mmol, 1.05 equiv.) were added. The mixture was heated at 30° C. for 16 hours. The reaction mixture was concentrated by Speedvac to give a residue that was purified by prep HPLC to give N-(5-(2-((3aR,5R,6aS)-2-(2,2,2-trifluoroethyl)octahydrocyclopenta [c]pyrrol-5-yl)ethoxy)-1H-indol-3-yl)spiro[2.3]hexane-1-carboxamide (34.18 mg, 0.072 mmol) as a powder. MS-ESI, 476.4 [M+H+]. 1H NMR (400 MHz, DMSO-d6) δ ppm 10.52 (s, 1H), 9.81 (s, 1H), 7.63 (d, 1H), 7.32 (d, 1H), 7.18 (d, 1H), 6.71 (dd, 1H), 3.97 (t, 2H), 3.18 (q, 2H), 2.64 (br d, 2H), 2.46 (br s, 1H), 2.42 (br d, 2H), 2.33-2.15 (m, 2H), 2.15-2.01 (m, 6H), 2.00-1.94 (m, 1H), 1.91-1.85 (m, 2H), 1.78 (q, 2H), 1.06 (t, 1H), 1.01-0.92 (m, 3H).


Example 30: Synthesis of 1-methyl-N-(5-(cis)-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 241)



embedded image


tert-butyl 3-((tert-butoxycarbonyl)amino)-5-((1S,3S)-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate (98.4 mg, 0.18 mmol, 1.0 equiv.) was dissolved in DCM (3 mL), then TFA (1 mL) was added to the solution. The mixture was heated at 30° C. for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and 1-methyl-1H-1,2,3-triazole-4-carboxylic acid (45.7 mg, 0.36 mmol, 2.0 equiv.) were dissolved in DMF (2 mL), then TEA (130 μl, 0.9 mmol, 5.0 equiv.) and HATU (71.8 mg, 0.189 mmol, 1.05 equiv.) were added. The mixture was heated at 30° C. for 16 hours. The reaction mixture was concentrated by Speedvac to give a residue that was purified by prep HPLC to give 1-methyl-N-(5-((1S,3S)-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (41.88 mg, 0.092 mmol) as a powder. MS-ESI, 456.3 [M+H+]. 1H NMR (400 MHz, DMSO-d6) δ ppm 10.78 (br s, 1H), 10.23 (s, 1H), 8.63 (s, 1H), 7.74-7.71 (m, 1H), 7.67 (d, 2H), 7.52 (d, 2H), 7.35 (d, 1H), 7.25 (d, 1H), 6.73 (dd, 1H), 4.72 (quin, 1H), 4.14 (s, 3H), 3.32-3.28 (m, 1H), 3.00 (q, 2H), 2.21-2.07 (m, 2H).


Example 31: Synthesis of 1-methyl-N-(5-((4-(trifluoromethyl)benzyl)oxy)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (Compound 240)



embedded image


tert-butyl 3-((tert-butoxycarbonyl)amino)-5-((4-(trifluoromethyl)benzyl)oxy)-1H-indole-1-carboxylate (91.08 mg, 0.18 mmol, 1.0 equiv.) was dissolved in DCM (3 mL), then TFA (1 mL) was added to the solution. The mixture was heated at 30° C. for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and 1-methyl-1H-1,2,3-triazole-4-carboxylic acid (45.7 mg, 0.36 mmol, 2.0 equiv.) were dissolved in DMF (2 mL), then TEA (130 μl, 0.9 mmol, 5.0 equiv.) and HATU (71.8 mg, 0.189 mmol, 1.05 equiv.) were added. The mixture was heated at 30° C. for 16 hours. The reaction mixture was concentrated by Speedvac to give a residue that was purified by prep HPLC to give 1-methyl-N-(5-((4-(trifluoromethyl)benzyl)oxy)-1H-indol-3-yl)-1H-1,2,3-triazole-4-carboxamide (41.88 mg, 0.092 mmol) as a powder. MS-ESI, 456.3 [M+H+]. 1H NMR (400 MHz, DMSO-d6) δ ppm 10.86-10.77 (m, 1H), 10.17 (s, 1H), 8.63 (s, 1H), 7.79-7.70 (m, 5H), 7.57 (d, 1H), 7.28 (d, 1H), 6.86 (dd, 1H), 5.22 (s, 2H), 4.14 (s, 3H).


Example 32: Synthesis of N-(5-((1R,3R)-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)thiazole-4-carboxamide (Compound 239)



embedded image


tert-butyl 3-((tert-butoxycarbonyl)amino)-5-((1R,3R)-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indole-1-carboxylate (98.3 mg, 0.18 mmol, 1.0 equiv.) was dissolved in DCM (3 mL), then TFA (1 mL) was added to the solution. The mixture was heated at 30° C. for 2 hours. The reaction mixture was concentrated by Speedvac to give a residue. Then the residue and thiazole-4-carboxylic acid (46.44 mg, 0.36 mmol, 2.0 equiv.) were dissolved in DMF (2 mL), then TEA (130 μl, 0.9 mmol, 5.0 equiv.) and HATU (71.8 mg, 0.189 mmol, 1.05 equiv.) were added. The mixture was heated at 30° C. for 16 hours. The reaction mixture was concentrated by Speedvac to give a residue that was purified by prep HPLC to give N-(5-((1R,3R)-3-(4-(trifluoromethyl)phenyl)cyclobutoxy)-1H-indol-3-yl)thiazole-4-carboxamide (30.8 mg, 0.067 mmol) as a powder. MS-ESI, 458.2 [M+H+]. 1H NM/R (400 MHz, DMSO-d6) δ ppm 10.82 (d, 1H), 10.09 (s, 1H), 9.27 (d, 1H), 8.43 (d, 1H), 7.78-7.67 (m, 3H), 7.59 (d, 2H), 7.27 (d, 1H), 7.18 (d, 1H), 6.76 (dd, 1H), 4.95 (quin, 1H), 3.85-3.76 (m, 1H), 2.69-2.58 (in, 4H).









TABLE 1







The compounds in Table 1 were prepared using the above procedure.















LC-MS,






MS-ESI,


Example
Compound





#
No.
Final compound
IUPAC Name
[M + H+].





33
222


embedded image


N-(5-{2- [(3aR,5R,6aS)-2- (2,2,2-trifluoro- ethyl)octahydro- cyclopenta[c]- pyrrol-5-yl]- ethoxy}-1H- indol-3-yl)spiro- [2.3]hexane-5- carboxamide
476.4





34
242


embedded image


N-(5-{2- [(3aR,5R,6aS)- 2-(2,2,2-trifluoro- ethyl)octahydro- cyclopenta[c]- pyrrol-5-yl]- ethoxy}-1H-indol- 3-yl)-1-methyl- 1H-1,2,3-triazole- 4-carboxamide
477.4





35
236


embedded image


N-(5-{[4-(tri- fluoromethyl)- phenyl]methoxy}- 1H-indol-3-yl)- spiro[2.3]hexane- 1-carboxamide
415.2





36
221


embedded image


N-(5-{[4-(tri- fluoromethyl)- phenyl]methoxy}- 1H-indol-3-yl)- spiro[2.3]hexane- 5-carboxamide
415.3





37
235


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}-1,3-thiazole- 5-carboxamide
458.2





38
234


embedded image


3,5-dimethyl-N- {5-[(1R,3R)-3-[4- (trifluoromethyl)- phenyl]cyclo- butoxy]-1H-indol- 3-yl}-1,2-oxazole- 4-carboxamide
470.3





39
233


embedded image


5-methyl-N-{5- [(1R,3R)-3-[4- (trifluoromethyl)- phenyl]cyclo- butoxy]-1H-indol- 3-yl}-1,2-oxazole- 4-carboxamide
456.3





40
232


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}-1,3-thiazole- 2-carboxamide
458.2





41
231


embedded image


3-fluoro-N-{5- [(1R,3R)-3-[4- (trifluoromethyl)- phenyl]cyclo- butoxy]-1H-indol- 3-yl}pyridine-2- carboxamide
470.2





42
212


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}bicyclo- [1.1.1]pentane-1- carboxamide
441.3





43
238


embedded image


3-methyl-N-{5- [(1R,3R)-3-[4- (trifluoromethyl)- phenyl]cyclo- butoxy]-1H-indol- 3-yl}bicyclo- [1.1.1]pentane-1- carboxamide
455.3





44
230


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}spiro[2.3]- hexane-1-carbox- amide
455.4





45
229


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}spiro[2.3]- hexane-5-carbox- amide
455.3





46
228


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}pyridine-2- carboxamide
452.3





47
227


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}pyridine-3- carboxamide
452.3





48
237


embedded image


1-methyl-N-{5- [(1R,3R)-3-[4- (trifluoromethyl)- phenyl]cyclo- butoxy]-1H-indol- 3-yl}-1H-pyrazole- 5-carboxamide
455.2





49
226


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}-1H-pyrazole- 5-carboxamide
441.3





50
225


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}pyridine-4- carboxamide
452.3





51
220


embedded image


1-methyl-N-{5- [(1R,3R)-3-[4-(tri- fluoromethyl)phen- yl]cyclobutoxy]- 1H-indol-3-yl}- 1H-imidazole-2- carboxamide
455.2





51
211


embedded image


N-{5-[(1R,3R)-3- [4-(trifluorometh- yl)phenyl]cyclo- butoxy]-1H-indol- 3-yl}-2-oxaspiro- [3.3]heptane-6- carboxamide
471.3









Biological Assays

STING pathway activation by the compounds described herein was measured using TTIP1-Dual™ cells (KO-IFNAR2).


TTIP1-Dual™ KO-IFNAR2 Cells (obtained from invivogen) were maintained in RPMI, 10% FCS, 5 ml P/S, 2 mM L-glut, 10 mM 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 μM. Cells were plated into the TC plates at 40 μL 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:

    • Solution A: 2 mL Optimem with one of the following stimuli:
      • 60 μL of 10 mM 2′3′cGAMP→150 μM stock
    • Solution B: 2 mL Optimem with 60 L 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 μL of transfection solution (A+B) was added on top of the plated cells, with a final 2′3′cGAMP concentration of 15 μM. 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 24 h. Luciferase reporter activity was then measured. EC50 values were calculated by using standard methods known in the art.


Luciferase reporter assay: 10 μL 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 μL of QLC Stabilizer per 25 mL of QUANTI-Luc™ Plus solution was added. 50 μL 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. EC50 values were calculated by using standard methods known in the art.


Table BA shows the activity of compounds in STING reporter assay: <0.008 μM=“++++++”; ≥0.008 and ≤0.04 μM=“+++++”; >0.04 and ≤0.2 μM=“++++”; >0.2 and <1 μM=“+++”≥1 and ≤5 μM=“++”; >5 and <100 μM=












TABLE BA








Pharmaron,




THP1_IFNAR2




STING 24 h




Luci




Normalized:



Compound
GeoMean



No.
EC50 (uM)









101
+++



102
++++



103
+++



104
+++



105
+++



106
+++



107
+++



108
+++



109
+++



110
+++



111
+



112
+++



113
+++



114
+++



115
+++



116
+++



117
+++



118
+



119
+++



120
+++



121
+++



122
++++



123
+++



124
++



125
++++



126
++++



127
+++



128
+++



129
+++



130
++++



131
+++



132
+++



133
++++



134
+++



135
+++



136
+++



137
+++



138
+++



139
+++



140
+++



141
+++



142
+++



143
++++



144
++++



145
+++



146
++++



147
+++



148
+++



149
+



150
+++



151
+++



152
+++



153
+++



154
+++



155
+++



156
+++



157
+++



158
++



159
+



160
+++



161
+++



162
+++



163
+++



164
+++



165
++



166
+++



167
++++



168
+++



169
++++



170
++



171
+++



172
+++



173
+++



174
+++



175
+++



176
+



177
++++



178
+++



179
+++



180
+++



181
+++



182
+++



183
++



184
+++



185
+++



186
+++



187
++



188
+++



189
+++



190
+++



191
++++



192
+++



193
+++



194
+++



195
+++



196
++



197
+++



198
+++



199
+++



200
+++



201
+++



202
+++



203
+++



204
++



205
+++



206
+++



207
+++



208
+++



209
++++



210
+++



211
++++



212
++++



213
+++



214
++++



215
+++



216
++++



217
+++



218
+++



219
+++



220
+++



221
+++



222
+++



223
+++



224
+++



225
+++



226
++++



227
+++



228
+++



229
++++



230
++++



231
++++



232
++++



233
+



234
+++



235
+++



236
++++



237
+++



238
++++



239
++++



240
++



241
+++



242
+++



243
+++



244
++++



245
+++



246
++++










Numbered Clauses

The compounds, compositions, methods, and other subject matter described herein are further described in the following numbered clauses:


1. A compound of Formula (I):




embedded image


or a pharmaceutically acceptable salt thereof or a tautomer thereof, wherein:


LA is -(L1)a1-(L2)a2-(L3)a3-(L4)a4-(L5)a5-*, wherein * represents the point of attachment to Q1;


a1, a2, a3, a4, and a5 are each independently 0 or 1, provided that a1+a2+a3+a4+a5≥1, and


each of L1, L3, and L5 is independently selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, S(O)0-2, and —C(═O)—;


provided that when one or both of a2 and a4 is 0, then the combinations of L1, L3, and L5 cannot form O—O, N—O, N—N, O—S, S—S, or N—S(O)0 bonds, and


each of L2 and L4 is independently selected from the group consisting of:

    • straight-chain C1-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb;
    • C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc; and
    • heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc;


Q1 is —Rg;


Y1, Y2, and Y3 are each independently selected from the group consisting of CR1, C(═O), N, and NR2;


X1 is selected from the group consisting of O, S, N, NR2, and CR1;


X2 is selected from the group consisting of O, S, N, NR4, and CR5;


each custom-character is independently a single bond or a double bond, provided that the five-membered ring comprising X1 and X2 is heteroaryl, and that the six-membered ring comprising Y1, Y2, and Y3 is aryl or heteroaryl;


further provided that LA cannot include a cyclic group directly attached to the 6-membered ring containing Y1, Y2, and Y3;


each occurrence of R1 and R5 is independently selected from the group consisting of: H; Rc; Rg; and -(Lg)bg-Rg;


each occurrence of R2 and R4 is independently selected from the group consisting of: H; Rd; Rg; and -(Lg)bg-Rg;


R6 is selected from the group consisting of: H; Rd; and Rg;


W is selected from the group consisting of:




embedded image


Ring B1 is a heteroarylene of 5 ring atoms, wherein 1-4 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S; wherein the heteroarylene of Ring B1 is optionally substituted with 1-2 substituents independently selected from the group consisting of oxo and Rc, provided that Ring B1 is attached to the C(═O)NR6 group via a ring carbon atom;


each LA is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-2 Ra; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa1 is 0, 1, or 2;


Ring C1 is selected from the group consisting of:

    • C3-12 cycloalkylene or C3-12 cycloalkenylene, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and (LAA)aa1-Rg;
    • heterocyclylene or heterocycloalkenylene of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and (LAA)aa1-Rg;
    • heteroarylene of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroarylene is optionally substituted with 1-4 substituents independently selected from the group consisting of Rc and (LAA)aa1Rg; and
    • C6-10 arylene optionally substituted with 1-4 substituents independently selected from the group consisting of Rc and (LAA)aa1-Rg;


R7 is selected from the group consisting of: Rg and -(L7)b7-Rg;


each L7 is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-2 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O); and


b7 is 1, 2, or 3;




embedded image


Ring B2 is a heteroarylene of 5 ring atoms, wherein 1-4 of the ring atoms are heteroatoms each independently selected from the group consisting of: N, NH, N(Rd), O, and S, wherein the heteroarylene of Ring B is optionally substituted with 1-2 substituents independently selected from the group consisting of: oxo and Rc, provided that Ring B is attached to the C(═O)NR6 group via a ring carbon atom;


each LAB is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa2 is 0, 1, 2, or 3;


Ring C2 is selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc;
    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; and
    • C6-10 aryl optionally substituted with 1-4 Rc


(iii) heteroaryl of 5 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc; provided the heteroaryl is attached to the C(═O)NR6 group via a ring carbon atom;




embedded image


P1, P2, P3, P4, and P5 are each independently selected from the group consisting of: N, NH, NRd, NR71, CH, CRC, CR71, and C(═O);


each occurrence of R71 is independently -(LAC)aa3-R8, wherein: each LAC is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra; —O—; —NRN; —S(O)0-2; C(O); C(O)O; OC(O); NRNC(O); C(O)NRN; NRNC(O)NRN; NRNC(O)O; and OC(O)NRN;


aa3 is 0, 1, 2, or 3;


each occurrence of R8 is independently Rg or C1-10 alkyl optionally substituted with 1-6 Ra1; and


each occurrence of RN is independently H or Rd;


(v) a bicyclic or polycyclic ring system selected from the group consisting of:

    • bicyclic or polycyclic C5-15 cycloalkyl or C5-15 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heterocyclyl or heterocycloalkenyl of 7-15 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heteroaryl of 8-15 ring atoms, wherein 1-6 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of: oxo, Rc, and -(LAD)bB-Rg; and
    • bicyclic or polycyclic C8-15 aryl optionally substituted with 1-4 substituents independently selected from the group consisting of: oxo, Rc, and -(LAD)bB-Rg,
    • provided the bicyclic or polycyclic heteroring is attached to the C(═O)NR6 group via a ring carbon atom;
    • each occurrence of LAD is selected from the group consisting of: —O—, —NH—, —NRd, —S(O)0-2, C(O), and C1-3 alkylene optionally substituted with 1-3 Ra; and
    • bB is 0, 1, 2, or 3;
    • AND




embedded image


LAE is selected from the group consisting of:

    • C1-6 alkylene, C2-6 alkenylene, or C2-6 alkynylene, each of which is optionally substituted with 1-6 Ra;
    • monocyclic C3-8 cycloalkylene or C3-8 cycloalkenylene, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; and
    • monocyclic heterocyclylene or heterocycloalkenylene of 3-8 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc, provided that the heterocycloylene or heterocycloalkenylene is attached to the C(═O)NR6 group via a ring carbon atom;


each LAF is independently selected from the group consisting of: C1-3 alkylene optionally substituted with 1-4 Ra1; —O—; —NH—; —NRd; —S(O)0-2; and C(O);


aa4 is 0, 1, 2, or 3; and


Ring C4 is Rg;


each occurrence of Ra and is independently selected from the group consisting of: —OH; -halo; —NReRf; C1-4 alkoxy; C1-4 haloalkoxy; —C(═O)O(C1-4 alkyl); —C(═O)(C1-4 alkyl); —C(═O)OH; —CONR′R″; —S(O)1-2NR′R″; —S(O)1-2(C1-4 alkyl); and cyano;


each occurrence of Rb and Rc is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy; C1-4 haloalkoxy; —S(O)1-2(C1-4 alkyl); —S(O)(═NH)(C1-4 alkyl); —NReRf; —OH; —S(O)1-2NR′R″; —C1-4 thioalkoxy; —NO2; —C(═O)(C1-10 alkyl); —C(═O)O(C1-4 alkyl); —C(═O)OH; —C(═O)NR′R″; and —SF5;


each occurrence of Rd is independently selected from the group consisting of: C1-6 alkyl optionally substituted with 1-3 independently selected Ra; —C(O)(C1-4 alkyl); —C(O)O(C1-4 alkyl); —CONR′R″; —S(O)1-2NR′R″; —S(O)1-2(C1-4 alkyl); —OH; and C1-4 alkoxy;


each occurrence of Rc and R is independently selected from the group consisting of: H; C1-6 alkyl optionally substituted with 1-3 substituents each independently selected from the group consisting of NR′R″, —OH, halo, C1-4 alkoxy, and C1-4 haloalkoxy; —C(O)(C1-4 alkyl); —C(O)O(C1-4 alkyl); —CONR′R″; —S(O)1-2NR′R″; —S(O)1-2(C1-4 alkyl); —OH; and C1-4 alkoxy;


each occurrence of Rg is independently selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;
    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh; and
    • C6-10 aryl optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rh;


each occurrence of Rh is independently selected from the group consisting of:

    • C3-12 cycloalkyl or C3-12 cycloalkenyl, each of which is optionally substituted with 1-4 R1;
    • heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 Ri;
    • heteroaryl of 5-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Ri; and
    • C6-10 aryl optionally substituted with 1-4 Ri;


each occurrence of Ri is independently selected from the group consisting of: C1-6 alkyl; C1-4 haloalkyl; C1-4 alkoxy; C1-4 haloalkoxy; and halo;


each occurrence of Lg is independently selected from the group consisting of: —O—, —NH—, —NRd, —S(O)0-2, C(O), and C1-3 alkylene optionally substituted with 1-3 Ra;


each occurrence of bg is independently 1, 2, or 3; and


each occurrence of R′ and R″ is independently selected from the group consisting of: H; —OH; and C1-4 alkyl.


2. The compound of clause 1, wherein a2 is 1.


3. The compound of clauses 1 or 2, wherein L2 is straight-chain C1-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb.


4. The compound of any one of clauses 1-3, wherein L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb.


5. The compound of any one of clauses 1-4, wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


6. The compound of any one of clauses 1-5, wherein L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—.


7. The compound of any one of clauses 1-6, wherein L2 is —CH2—.


8. The compound of any one of clauses 1-4, wherein L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


9. The compound of any one of clauses 1-4 or 8, wherein L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb.


10. The compound of any one of clauses 1-4 or 8-9, wherein L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -(L3)a3-.


11. The compound of any one of clauses 1-4 or 8-10, wherein L2 is —CH2CH2—.


12. The compound of any one of clauses 1-4 or 8, wherein L2 is straight-chain C3 alkylene which is optionally substituted with 1-3 Rb.


13. The compound of any one of clauses 1-4, 8, or 12, wherein L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents point of attachment to -(L3)a3-.


14. The compound of any one of clauses 1-3, wherein L2 is straight-chain C2-6 alkenylene, which is optionally substituted with 1-6 Rb.


15. The compound of any one of clauses 1-3 or 14, wherein L2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 Rb.


16. The compound of any one of clauses 1-3 or 14-15, wherein L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents the point of attachment to -(L3)a3-.


17. The compound of clauses 1 or 2, wherein L2 is selected from the group consisting of:

    • C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc; and
    • heterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc.


18. The compound of any one of clauses 1-2 or 17, wherein L2 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


19. The compound of any one of clauses 1-2 or 17-18, wherein L2 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0, 1, or 2; Q2 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L3)a3-.


20. The compound of clause 19, wherein Q2 is CH.


21. The compound of clauses 19 or 20, wherein n1 and n2 are each 0.


22. The compound of any one of clauses 1-2 or 17-21, wherein L2 is




embedded image


wherein the asterisk represents the point of attachment to -(L3)a3-.


23. The compound of clause 1, wherein a2 is 0.


24. The compound of any one of clauses 1-23, wherein a1 is 1.


25. The compound of any one of clauses 1-24, wherein L1 is selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, and —S—.


26. The compound of any one of clauses 1-25, wherein L1 is —O—.


27. The compound of any one of clauses 1-23, wherein a1 is 0.


28. The compound of any one of clauses 1-27, wherein a3 is 1.


29. The compound of any one of clauses 1-28, wherein L3 is selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, and —S—


30. The compound of any one of clauses 1-29, wherein L3 is —O—.


31. The compound of any one of clauses 1-29, wherein L3 is —N(H)— or —N(Rd)—, optionally —N(H)—.


32. The compound of any one of clauses 1-27, wherein a3 is 0.


33. The compound of any one of clauses 1-32, wherein a4 is 1.


34. The compound of any one of clauses 1-33, wherein L4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


35. The compound of any one of clauses 1-34, wherein L4 is —CH2—.


36. The compound of any one of clauses 1-33, wherein L4 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


37. The compound of any one of clauses 1-33 or 36, wherein L4 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n3 and n4 are independently 0, 1, or 2; Q3 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L5)a5-.


38. The compound of clause 37, wherein n3 and n4 are each 1.


39. The compound of clauses 37 or 38, wherein Q3 is N.


40. The compound of any one of clauses 1-33 or 36-39, wherein L4 is




embedded image


wherein the asterisk represents the point of attachment to -(L5)a5-.


41. The compound of any one of clauses 1-32, wherein a4 is 0.


42. The compound of any one of clauses 1-41, wherein a5 is 0.


43. The compound of clause 1, wherein one of a1, a3, and a5 is 1, and the other two are 0.


44. The compound of clauses 1 or 43, wherein one of a2 and a4 is 1, and the other is 0 or 1.


45. The compound of any one of clauses 1 or 43-44, wherein a1 and a2 are each 1.


46. The compound of any one of clauses 1 or 43-45, wherein:


a1 and a2 are each 1;


L1 is —O—, —N(H)—, or —N(Rd)—;


L2 is selected from the group consisting of:

    • straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb;
    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


47. The compound of any one of clauses 1 or 43-46, wherein:


a1 and a2 are each 1;


L1 is —O—; and


L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


48. The compound of any one of clauses 1 or 43-47, wherein:


a1 and a2 are each 1;


L1 is —O—; and


L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—.


49. The compound of any one of clauses 1 or 43-47, wherein:


a1 and a2 are each 1;


L1 is —O—; and


L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


50. The compound of clause 49, wherein L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb.


51. The compound of clauses 49 or 50, wherein L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -(L3)a3-.


52. The compound of any one of clauses 49-51, wherein L2 is —CH2CH2—.


53. The compound of any one of clauses 1 or 43-46, wherein:


a1 and a2 are each 1;


L1 is —O—;


L2 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


54. The compound of clause 53, wherein L2 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0, 1, or 2; Q2 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L3)a3-.


55. The compound of clause 54, wherein n1 and n2 are independently 0 or 1, optionally 0; and Q2 is CH; optionally wherein n1 and n2 are 0 and Q2 is CH; optionally wherein L2 is cyclobutane-diyl optionally substituted with 1-2 Rc; optionally wherein L2 is cyclobutane-1,3-diyl optionally substituted with 1-2 Rc; optionally wherein L2 is unsubstituted cyclobutane-diyl; optionally wherein L2 is unsubstituted cyclobutane-1,3-diyl.


56. The compound of any one of clauses 43-55, wherein a3, a4, and a5 are each 0, optionally wherein LA is —O—CH2CH2—*, or




embedded image


(such as or




embedded image


wherein * represents the point of attachment to Q1.


57. The compound of any one of clauses 43-55, wherein a3 and a5 are 0; and a4 is 1.


58. The compound of clause 57, wherein L4 is selected from the group consisting of:

    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


59. The compound of clauses 57 or 58, wherein L4




embedded image


which is optionally substituted with 1-2 Rc, wherein n3 and n4 are independently 0, 1, or 2; Q3 is CH, CRc, or N; and the asterisk represents the point of attachment to -(L5)a5-.


60. The compound of clause 59, wherein n3 and n4 are independently 0 or 1; and Q3 is N.


61. The compound of any one of clauses 1 or 43-44, wherein: a1 is 0; and a2 is 1.


62. The compound of any one of clauses 1, 43-44, or 61, wherein a1 is 0; a2 is 1; and L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb.


63. The compound of clauses 61 or 62, wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


64. The compound of any one of clauses 61-63, wherein L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—.


65. The compound of any one of clauses 61-64, wherein L2 is —CH2—.


66. The compound of any one of clauses 61-63, wherein L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


67. The compound of any one of clauses 61-63 or 66, wherein L2 is straight-chain C2 alkylene, which is optionally substituted with 1-3 Rb.


68. The compound of any one of clauses 61-63 or 66-67, wherein L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -(L3)a3-.


69. The compound of any one of clauses 61-63 or 66-68, wherein L2 is —CH2CH2—.


70. The compound of any one of clauses 61-63 or 66, wherein L2 is straight-chain C3 alkylene, which is optionally substituted with 1-3 Rb.


71. The compound of any one of clauses 61-63, 66, or 70, wherein L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents point of attachment to -(L3)a3-.


72. The compound of any one of clauses 61-71, wherein a3 is 0; a4 is 0; and a5 is 0.


73. The compound of any one of clauses 61-71, wherein a3 is 1.


74. The compound of clause 73, wherein a3 is 1; and L3 is selected from the group consisting of: is —O—, —N(H)—, and —N(Rd)—.


75. The compound of clauses 73 or 74, wherein a3 is 1; and L3 is —O—.


76. The compound of any one of clauses 61-71 or 73-74, wherein a3 is 1; and L3 is —N(H)— or —N(Rd)—, optionally —N(H)—.


77. The compounds of any one of clauses 61-71 or 73-76, wherein a4 is 1; and L4 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


78. The compound of any one of clauses 61-71 or 73-77, wherein a4 is 1; and L4 is —CH2—.


79. The compound of any one of clauses 61-71 or 73-77, wherein a4 is 0; and a5 is 0, optionally wherein LA is —CH2CH2—O—*, wherein * represents to point of attachment to Q1.


80. The compound of clause 1, wherein a1 is 0; a2 is 1; L2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 Rb.


81. The compound of clause 80, wherein L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents the point of attachment to -(L3)a3-.


82. The compound of clauses 80 or 81, wherein a3 is 0; a4 is 0; and a5 is 0.


83. The compound of any one of clauses 1-82, wherein Q1 is selected from the group consisting of:

    • heteroaryl of 5-12 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-4 Rc′; and
    • C6-10 aryl optionally substituted with 1-4 Rc′.


84. The compound of any one of clauses 1-82, wherein Q1 is selected from the group consisting of:

    • heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


85. The compound of any one of clauses 1-82, wherein Q1 is selected from the group consisting of:

    • heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


86. The compound of any one of clauses 1-85, wherein Q1 is phenyl optionally substituted with 1-3 Rc′.


87. The compound of any one of clauses 1-86, wherein Q1 is selected from the group consisting of:




embedded image


88. The compound of any one of clauses 1-85, wherein Q1 is heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 Rc′.


89. The compound of any one of clauses 1-85 or 88, wherein Q1 is pyridyl, which is optionally substituted with 1-3 Rc′.


90. The compound of any one of clauses 1-85 or 88-89, wherein Q1 is selected from the group consisting of:




embedded image


91. The compound of any one of clauses 1-82, wherein Q1 is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc′.


92. The compound of any one of clauses 1-82 or 91, wherein Q1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R1.


93. The compound of any one of clauses 1-82 or 91-92, wherein Q1 is heterocyclyl of 4-8 ring atoms, wherein 1-2 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, provided that one ring atom is N(Rd),


and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and R1.


94. The compound of any one of clauses 1-82 or 91-93, wherein Q1 is




embedded image


wherein m1 and m2 are each independently 0, 1, or 2; and wherein Q1 is optionally substituted with 1-2 Rc′.


95. The compound of any one of clauses 1-82 or 91-94, wherein Q1 is




embedded image


96. The compound of any one of clauses 1-82 or 91-94, wherein Q1 is




embedded image


97. The compound any one of clauses 91-96, wherein each Rd present in Q1 is independently selected from the group consisting of: —C(O)O(C1-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected Ra.


98. The compound of any one of clauses 91-97, wherein each Rd present in Q1 is C1-6 alkyl optionally substituted with 1-3 independently selected halo.


99. The compound of any one of clauses 91-98, wherein each Rd present in Q1 is:


i. C1-4 alkyl substituted with 1-3 —F;


ii. C2-3 alkyl substituted with 1-3 —F; or


iii. —CH2CF3.


100. The compound of any one of clauses 83-99, wherein each Rc present in Q1 is independently selected from the group consisting of: halo; cyano; C1-4 alkoxy; C1-4 haloalkoxy; and C1-10 alkyl which is optionally substituted with 1-6 independently selected Ra.


101. The compound of any one of clauses 83-100, wherein each Rc present in Q1 is independently selected from the group consisting of: halo; cyano; C1-4 alkoxy; C1-4 haloalkoxy; and C1-6 alkyl which is optionally substituted with 1-6 independently selected halo.


102. The compound of any one of clauses 83-101, wherein each Rc present in Q1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo.


103. The compound of any one of clauses 83-102, wherein each Rc present in Q1 is:


i. C1-3 alkyl which is optionally substituted with 1-6 —F; or


ii. CF3.


104. The compound of any one of clauses 83-102, wherein each Rc present in Q1 is an independently selected halo, optionally —F or —Cl;


105. The compound of any one of clauses 1-104, wherein Y1 is CR1.


106. The compound of any one of clauses 1-105, wherein Y2 is CR1.


107. The compound of any one of clauses 1-106, wherein Y3 is CR1.


108. The compound of any one of clauses 1-107, wherein each occurrence of R1 is independently H or Rc.


109. The compound of any one of clauses 1-108, wherein each occurrence of R1 is H.


110. The compound of any one of clauses 1-108, wherein 1-2 occurrence of R1 is Rc; and each remaining occurrence of R1 is H.


111. The compound of any one of clauses 1-108 or 110, wherein one occurrence of R1 is halo, optionally —F or —C1; and each remaining occurrence of R1 is H.


112. The compound of any one of clauses 1-111, wherein Y1, Y2, and Y3 are each independently selected CR1.


113. The compound of any one of clauses 1-107 or 112, wherein Y1, Y2, and Y3 are each CH.


114. The compound of any one of clauses 1-107 or 112, wherein one of Y1, Y2, and Y3 is CRc, optionally C-halo; and each of the remaining two Y1, Y2, and Y3 is CH.


115. The compound of any one of clauses 1-114, wherein X1 is NR2.


116. The compound of any one of clauses 1-115, wherein X1 is NH.


117. The compound of any one of clauses 1-116, wherein X2 is CR5.


118. The compound of any one of clauses 1-117, wherein X2 is CH.


119. The compound of any one of clauses 1-114, wherein X1 is NR2; and X2 is CR5.


120. The compound of any one of clauses 1-114 or 119, wherein X1 is NH; and X2 is CH.


121. The compound of any one of clauses 1-104, wherein Y, Y2, and Y3 are each an independently selected CR1; X1 is NR2; and X2 is CR5.


122. The compound of any one of clauses 1-104 or 121, wherein Y, Y2, and Y3 are each CH; X1 is NH; and X2 is CH.


123. The compound of any one of clauses 1-122, wherein R6 is H.


124. The compound of any one of clauses 1-123, wherein W has formula (A-1).


125. The compound of any one of clauses 1-123, wherein W has formula (A-2).


126. The compound of any one of clauses 1-123, wherein W has formula (A-3-1).


127. The compound of any one of clauses 1-123, wherein W has formula (A-4).


128. The compound of any one of clauses 1-123, wherein W is defined according to (i).


129. The compound of any one of clauses 1-123, wherein W is defined according to (ii).


130. The compound of any one of clauses 1-123, wherein W is defined according to (iii).


131. The compound of any one of clauses 1-123, wherein W is defined according to (iv).


132. The compound of any one of clauses 1-123, wherein W is defined according to (v).


133. The compound of any one of clauses 1-123, wherein W is defined according to (vi).


134. The compound of any one of clauses 1-123, wherein W has the formula:




embedded image


wherein n7 is 0, 1, or 2; and each Rc7 is an independently selected Rc, such as:




embedded image


135. The compound of any one of clauses 1-123, wherein W is heteroaryl of 5 ring atoms, such as thienyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl, or tetrazolyl, e.g., thiazolyl.


136. The compound of any one of clauses 1-123, wherein W is selected from the group consisting of:

    • bicyclic or polycyclic C5-15 cycloalkyl or C5-15 cycloalkenyl, each optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;
    • bicyclic or polycyclic heterocyclyl or heterocycloalkenyl of 7-15 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo, Rc, and -(LAD)bB-Rg;


137. The compound of any one of clauses 1-123, wherein W is unsubstituted bicyclic C5-15 cycloalkyl.


138. The compound of any one of clauses 1-123, wherein W is C1-C6 alkyl substituted with 1-6 R9.


139. The compound of any one of clauses 1-123 or 134-138, wherein W is




embedded image


140. The compound of any one of clauses 1-123, wherein W is




embedded image


141. The compound of clause 1, wherein the compound is a compound of Formula (I-a):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L1 is selected from the group consisting of: —O—, —N(H)—, and —N(Rd)—;


L2 is selected from the group consisting of:

    • straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb;
    • C3-8 cycloalkylene, which is optionally substituted with 1-3 Rc; and
    • heterocyclylene having 4-8 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene is optionally substituted with 1-3 Rc.


142. The compound of clause 141, wherein L1 is —O—.


143. The compound of clauses 141 or 142, wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.


144. The compound of any one of clauses 141-143, wherein L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2—, optionally wherein L2 is —CH2—.


145. The compound of any one of clauses 141-143 wherein L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb.


146. The compound of any one of clauses 141-143 or 145, wherein L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -Q1.


147. The compound of clause 146, wherein L2 is —CH2CH2—.


148. The compound of any one of clauses 141-143, wherein L2 is straight-chain C3 alkylene which is optionally substituted with 1-3 Rb.


149. The compound of clauses 141 or 142, wherein L2 is:




embedded image


which is optionally substituted with 1-2 Rc, wherein n1 and n2 are independently 0, 1, or 2; Q2 is CH, CRc, or N; and the asterisk represents the point of attachment to Q1;


150. The compound of clause 149, wherein n1 and n2 are independently 0 or 1, optionally 0; and Q2 is CH; optionally wherein n1 and n2 are 0 and Q2 is CH; optionally wherein L2 is cyclobutane-diyl optionally substituted with 1-2 Rc; optionally wherein L2 is cyclobutane-1,3-diyl optionally substituted with 1-2 Rc; optionally wherein L2 is cyclobutane-diyl optionally substituted with 1-2 Rc; optionally wherein L2 is unsubstituted cyclobutane-diyl; optionally wherein L2 is unsubstituted cyclobutane-1,3-diyl.


151. The compound of clause 141, wherein L1 is —O—; and L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


152. The compound of clause 151, wherein L2 is:


i. straight-chain C2 alkylene which is optionally substituted with 1-3 Rb;


ii. selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -Q1; or


iii. —CH2CH2—.


153. The compound of clause 141, wherein L1 is —O—; and L2 is:


i. selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2; or


ii. —CH2—.


154. The compound of clause 1, wherein the compound is a compound of Formula (I-b):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L2 is straight-chain C1-6 alkylene or straight-chain C2-6 alkenylene, each of which is optionally substituted with 1-6 Rb.


155. The compound of clause 154, wherein L2 is straight-chain C2-3 alkylene which is optionally substituted with 1-3 Rb.


156. The compound of clauses 154 or 155, wherein L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb.


157. The compound of any one of clauses 154-156, wherein L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -Q1, optionally wherein L2 is —CH2CH2—.


158. The compound of clauses 154-155, wherein L2 is straight-chain C3 alkylene which is optionally substituted with 1-3 Rb.


159. The compound of any one of clauses 154-155 or 158, wherein L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents point of attachment to -Q1, optionally wherein L2 is




embedded image


160. The compound of clause 154, wherein L2 is straight-chain C2-4 alkenylene, which is optionally substituted with 1-3 Rb.


161. The compound of clauses 154 or 160, wherein L2 is selected from the group consisting of:




embedded image


wherein the asterisk represents the point of attachment to -Q1.


162. The compound of clause 1, wherein the compound is a compound of Formula (I-c):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L2 and L4 are independently selected straight-chain C1-3 alkylene which is optionally substituted with 1-6 Rb; and


L3 is selected from the group consisting of: —O—, —N(H)—, and —N(Rd)—.


163. The compound of clause 162, wherein L2 and L4 are independently selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2.


164. The compound of clauses 162 or 163, wherein L2 and L4 are each —CH2—.


165. The compound of any one of clauses 162-164, wherein L3 is —O—.


166. The compound of any one of clauses 162-164, wherein L3 is —N(H)— or —N(Rd)—, optionally —N(H)—.


167. The compound of clause 1, wherein the compound is a compound of Formula (I-d):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


L2 is straight-chain C1-3 alkylene which is optionally substituted with 1-6 Rb; and


L3 is selected from the group consisting of: —O—, —N(H)—, and —N(Rd)—.


168. The compound of clause 167, wherein L2 is selected from the group consisting of: —CH2—, —CHRb—, and —C(Rb)2.


169. The compound of clause 167, wherein L2 is straight-chain C2 alkylene which is optionally substituted with 1-3 Rb.


170. The compound of clauses 167 or 169, wherein L2 is selected from the group consisting of: —CH2CH2—, —CH2CH(Rb)—*, and —CH2C(Rb)2—*, wherein the asterisk represents point of attachment to -L3, optionally wherein L2 is —CH2CH2—.


171. The compound of any one of clauses 167-170, wherein L3 is —O—.


172. The compound of any one of clauses 167-170, wherein L3 is —N(H)— or —N(Rd)—, optionally —N(H)—.


173. The compound of any one of clauses 141-172, wherein Q1 is selected from the group consisting of:

    • heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


174. The compound of any one of clauses 141-173, wherein Q1 is selected from the group consisting of:

    • heteroaryl of 6 ring atoms, wherein 1-2 ring atoms are ring nitrogen atoms, and wherein the heteroaryl is optionally substituted with 1-3 Rc′; and
    • phenyl optionally substituted with 1-3 Rc′.


175. The compound of any one of clauses 141-174, wherein Q1 is:


i. phenyl or pyridyl, each optionally substituted with 1-3 Rc′;


ii.




embedded image


iii. any groups of i or ii, wherein each Rc present in Q1 is independently selected from the group consisting of: halo and C1-3 alkyl which is optionally substituted with 1-6 independently selected halo; or


iv. any groups of i or ii, wherein each Rc present in Q1 is independently selected from the group consisting of: —F, —Cl, and —CF3.


176. The compound of any one of clauses 141-172, wherein Q1 is heterocyclyl of 4-10 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc′.


177. The compound of any one of clauses 141-172 or 176, wherein Q1 is:




embedded image


wherein m1 and m2 are each independently 0, 1, or 2;




embedded image


iii any groups of i or ii, wherein the Rd present in Q1 is selected from the group consisting of: —C(O)O(C1-4 alkyl); and C1-6 alkyl optionally substituted with 1-3 independently selected Ra; or


iv. any groups of i or ii, wherein the Rd present in Q1 is C2-3 alkyl substituted with 1-3 —F.


178. The compound of any one of clauses 141-177, wherein each R1 is H.


179. The compound of any one of clauses 141-177, wherein one occurrence of R1 is Rc; and each remaining R1 is H.


180. The compound of any one of clauses 141-179, wherein R2 is H; and R5 is H.


181. The compound of any one of clauses 141-180, wherein W has formula (A-1).


182. The compound of any one of clauses 141-180, wherein W has formula (A-2).


183. The compound of any one of clauses 141-180, wherein W has formula (A-3-1).


184. The compound of any one of clauses 141-180, wherein W has formula (A-4).


185. The compound of any one of clauses 141-180, wherein W is defined according to (iii).


186. The compound of any one of clauses 141-180, wherein W is defined according to (iv).


187. The compound of any one of clauses 141-180, wherein W is defined according to (v).


188. The compound of any one of clauses 141-180, wherein W is defined according to (vi).


189. The compound of clause 1, wherein the compound is selected from the group consisting of compounds delineated in Table C1, and a pharmaceutically acceptable salt thereof.


190. A pharmaceutical composition comprising a compound of clauses 1-189 and one or more pharmaceutically acceptable excipients.


191. A method for inhibiting STING activity, the method comprising contacting STING with a compound or a pharmaceutically acceptable salt thereof as defined in any one of clauses 1-126; or a pharmaceutical composition as defined in clause 190.


192. The method of clause 191, wherein the inhibiting comprises antagonizing STING.


193. The method of any one of clauses 191-192, which is carried out in vitro.


194. The method of clauses 193, wherein the method comprises contacting a sample comprising one or more cells comprising STING with the compound.


195. The method of clauses 193 or 194, wherein the one or more cells are one or more cancer cells.


196. The method of clauses 194 or 195, 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.


197. The method of clauses 191 or 192, which is carried out in vivo.


198. The method of clause 197, 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.


199. The method of clause 198, wherein the subject is a human. 200. The method of clause 199, wherein the disease is cancer.


201. The method of clause 200, 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.


202. The method of clauses 200 or 201, wherein the cancer is a refractory cancer.


203. The method of clause 198, wherein the compound is administered in combination with one or more additional cancer therapies.


204. The method of clause 203, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.


205. The method of clause 204, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.


206. The method of clause 205, 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 topoisomerase; 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, Muromonab-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-β (TGFβ), 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, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD160, HVEM LIGHT, HVEM-BTLA-CD160, 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 PD1 or PD-L1).


207. The method of any one of clauses 198-206, wherein the compound is administered intratumorally.


208. A method of treating cancer, comprising administering to a subject in need of such treatment an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.


209. The method of clause 208, 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.


210. The method of clause 208 or 209, wherein the cancer is a refractory cancer.


211. The method of clause 208, wherein the compound is administered in combination with one or more additional cancer therapies.


212. The method of clause 211, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.


213. The method of clause 212, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.


214. The method of clause 212, 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 topoisomerase; 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, Muromonab-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-β (TGFβ), 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, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD160, HVEM LIGHT, HVEM-BTLA-CD160, 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 PD1 or PD-L1).


215. The method of any one of clauses 208-214, wherein the compound is administered intratumorally.


216. A method of inducing an immune response in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.


217. The method of clause 216, wherein the subject has cancer.


218. The method of clause 217, wherein the subject has undergone and/or is undergoing and/or will undergo one or more cancer therapies.


219. The method of clause 217, 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.


220. The method of clause any one of clauses 217-219, wherein the cancer is a refractory cancer.


221. The method of clause 219, wherein the immune response is an innate immune response.


222. The method of clause 221, wherein the at least one or more cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.


223. The method of clause 222, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.


224. The method of clause 223, 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 topoisomerase; 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, Muromonab-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-β (TGFβ), 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, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD160, HVEM LIGHT, HVEM-BTLA-CD160, 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 PD1 or PD-L1).


225. A method of treatment of a disease in which increased (e.g., excessive) STING 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 as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.


226. A method of treatment comprising administering 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 an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.


227. A method of treatment comprising administering to a subject a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190, wherein the compound or composition is administered in an amount effective to treat a disease in which increased (e.g., excessive) STING signaling contributes to the pathology and/or symptoms and/or progression of the disease, thereby treating the disease.


228. The method of any one of clauses 225-227, wherein the disease is cancer.


229. The method of clause 228, 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.


230. The method of clause 228 or 229, wherein the cancer is a refractory cancer.


231. The method of any one of clauses 228-230, wherein the compound is administered in combination with one or more additional cancer therapies.


232. The method of clause 231, wherein the one or more additional cancer therapies comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.


233. The method of clause 232, wherein chemotherapy comprises administering one or more additional chemotherapeutic agents.


234. The method of clause 233, 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 topoisomerase; 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, Muromonab-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-β (TGFβ), 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, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD160, HVEM-LIGHT, HVEM-BTLA-CD160, 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 PD1 or PD-L1).


235. The method of any one of clauses 225-234, wherein the compound is administered intratumorally.


236. A method of treatment of a disease, disorder, or condition associated with STING, comprising administering to a subject in need of such treatment an effective amount of a compound as defined in any one of clauses 1-189, or a pharmaceutical composition as defined in clause 190.


237. The method of clause 236, 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.


238. The method of clause 237, wherein the disease, disorder, or condition is a type I interferonopathy (e.g., STING-associated vasculopathy with onset in infancy (SAVI)).


239. The method of clause 238, wherein the type I interferonopathy is STING-associated vasculopathy with onset in infancy (SAVI)).


240. The method of clause 237, wherein the disease, disorder, or condition is Aicardi-Goutieres Syndrome (AGS).


241. The method of clause 237, wherein the disease, disorder, or condition is a genetic form of lupus.


242. The method of clause 237, wherein the disease, disorder, or condition is inflammation-associated disorder.


243. The method of clause 242, wherein the inflammation-associated disorder is systemic lupus erythematosus.


244. The method of any one of clauses 191-243, wherein the method further comprises identifying the subject.


245. A combination comprising a compounds defined in any one of clauses 1 to 126 or a pharmaceutically acceptable salt or tautomer thereof, and one or more therapeutically active agents.


246. A compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 190, for use as a medicament.


247. A compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 190, for use in the treatment of a disease, condition or disorder modulated by STING inhibition.


248. A compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or the pharmaceutical composition defined in clause 190, for use in the treatment of a disease mentioned in any one of clauses 191-244.


249. Use of a compound defined in any one of clauses 1-189 or a pharmaceutically acceptable salt or tautomer thereof, or a pharmaceutical composition defined in clause 190, in the manufacture of a medicament for the treatment of a disease mentioned in in any one of clauses 191-244.

Claims
  • 1. A compound of Formula (I):
  • 2. The compound of claim 1, wherein a2 is 1.
  • 3. The compound of claim 1, wherein L2 is straight-chain C1-6 alkylene, straight-chain C2-6 alkenylene, or straight-chain C2-6 alkynylene, each of which is optionally substituted with 1-6 Rb, optionally wherein L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb;optionally wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.
  • 4. The compound of claim 1, wherein L2 is selected from the group consisting of: C3-10 cycloalkylene or C3-10 cycloalkenylene, each of which is optionally substituted with 1-3 Rc; andheterocyclylene or heterocycloalkenylene, each having 4-10 ring atoms wherein 1-3 ring atoms are ring heteroatoms each independently selected from the group consisting of: N, N(H), N(Rd), O, and S(O)0-2, wherein the heterocyclylene or heterocycloalkenylene is optionally substituted with 1-3 Rc.
  • 5. The compound of claim 1, wherein at is 1.
  • 6. The compound of claim 1, wherein L1 is selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, and —S—, optionally wherein L1 is —O—.
  • 7. The compound of claim 1, wherein a1 is 0.
  • 8. The compound of claim 1, wherein a3 is 1.
  • 9. The compound of claim 1, wherein L3 is selected from the group consisting of: —O—, —N(H)—, —N(Rd)—, and —S—, optionally wherein L3 is —O—.
  • 10. The compound of claim 1, wherein a3 is 0.
  • 11. The compound of claim 1, wherein a4 is 1.
  • 12. The compound of claim 1, wherein: a1 and a2 are each 1;
  • 13. The compound of claim 12, wherein a3, a4, and a5 are each 0, optionally wherein LA is —O—CH2CH2—*, or
  • 14. The compound of claim 1, wherein a1 is 0; a2 is 1; optionally wherein L2 is straight-chain C1-6 alkylene, which is optionally substituted with 1-6 Rb, optionally wherein L2 is straight-chain C1-3 alkylene, which is optionally substituted with 1-3 Rb.
  • 15. The compound of claim 14, wherein a3 is 1; optionally, wherein L3 is selected from the group consisting of: is —O—, —N(H)—, and —N(Rd)—, optionally wherein L3 is —O—.
  • 16. The compound of claim 14, wherein a4 is 0; and a5 is 0, optionally wherein LA is —CH2CH2—O—*, wherein * represents to point of attachment to Q1.
  • 17. The compound of claim 1, wherein Q1 is selected from the group consisting of: heteroaryl of 5-6 ring atoms, wherein 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heteroaryl is optionally substituted with 1-3 Rc; andphenyl optionally substituted with 1-3 Rc.
  • 18. The compound of claim 1, wherein Q1 is heterocyclyl or heterocycloalkenyl of 3-12 ring atoms, wherein 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(O)0-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with 1-4 substituents independently selected from the group consisting of oxo and Rc; optionally wherein Q1 is
  • 19. The compound of claim 1, wherein Y1 is CR1; Y2 is CR1; and/or Y3 is CR1.
  • 20. The compound of claim 1, wherein X1 is NR2; and X2 is CR5; optionally wherein X1 is NH; and X2 is CH.
  • 21. The compound of claim 1, wherein R6 is H.
  • 22. The compound of claim 1, wherein W has formula (A-1) (A-2), or (A-4); optionally wherein W has formula (A-1); optionally wherein W has formula (A-2); optionally wherein W has formula (A-4).
  • 23. The compound of claim 1, wherein W is defined according to (iii), (iv), or (v); optionally wherein W is defined according to (iii); optionally wherein W is defined according to (iv); optionally wherein W is defined according to (v).
  • 24. The compound of claim 1, wherein the compound is selected from the group consisting of the compounds delineated in Table C1 or a pharmaceutically acceptable salt thereof.
  • 25. A pharmaceutical composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof.
  • 26. A method for inhibiting STING activity, the method comprising contacting STING with a compound as claimed in claim 1, or a pharmaceutically acceptable salt thereof.
  • 27. A method of inducing an immune response in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound as claimed in claim 1, or a pharmaceutically acceptable salt thereof.
  • 28. A method of treatment of disease, disorder, or condition associated with STING, such as a disease, disorder, or condition, in which increased STING signaling, such as excessive STING signaling, contributes to the pathology and/or symptoms and/or progression of the disease, such as cancer, comprising administering to a subject in need of such treatment an effective amount of a compound as claimed in claim 1, or a pharmaceutically acceptable salt thereof.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application Ser. No. 63/298,987, filed on Jan. 12, 2022, which is incorporated herein by reference in its entirety.

Provisional Applications (1)
Number Date Country
63298987 Jan 2022 US