Indoles, Indazoles, and Related Analogs for Inhibiting YAP/TAZ-TEAD

Abstract
The present disclosure relates to novel compounds, to said compounds for use as a medicine, more in particular for the prevention or treatment of diseases mediated by activity of YAP/TAZ-TEAD transcription, yet more in particular for the prevention or treatment of cancer or fibrosis. The present disclosure also relates to a method for the prevention or treatment of said diseases comprising the use of the novel compounds.
Description
FIELD

The present disclosure relates to novel compounds. The present disclosure also relates to the compounds for use as a medicine, more in particular for the prevention or treatment of diseases mediated by activity of YAP/TAZ-TEAD transcription, such as for the prevention or treatment of cancer or fibrosis. Methods for the prevention or treatment of the diseases comprising the use of the novel compounds are also disclosed herein.


The present disclosure furthermore relates to pharmaceutical compositions or combination preparations of the novel compounds, as well as to the compositions or preparations for use as a medicine, for example for the prevention or treatment of diseases mediated by activity of YAP/TAZ-TEAD transcription such as the prevention or treatment of cancer or fibrosis. Processes for the preparation of the compounds are also disclosed herein.


BACKGROUND

Hippo signaling is critical to restrict organ size through inactivation of the YAP/TAZ-TEAD transcriptional complex. In several aggressive solid cancers, Hippo signaling is inactivated through loss-of-function mutations or deletions in the genes encoding the upstream regulators (e.g. NF2, MST1/2 or LATS1/2), unleashing constitutive YAP/TAZ-TEAD transcriptional activity leading to unbridled tumor growth and metastasis. Knock-out, knockdown or pharmacologic inactivation of YAP/TAZ-TEAD is sufficient to impair YAP/TAZ-dependent tumorigenesis. The YAP/TAZ-TEAD complex can be pharmacologically inactivated through targeted disruption of the YAP/TAZ-TEAD protein-protein interaction interface, or through an allosteric autopalmitoylation pocket in TEAD.


The main physiologic function of the Hippo pathway is to restrict tissue growth in adult tissue and modulate cell proliferation, differentiation and migration in developing organs. The core of the Hippo pathway consists of a kinase cascade, transcription coactivators and DNA-binding partners. In mammals, the Ste20-like kinases, MST1/2 (homologs of Drosophila Hippo) phosphorylate and activate Large Tumor Suppressor 1/2 (LATS1/2). NF2 is a scaffold for the core Hippo kinases, promoting LATS1/2 activation by tethering MST1/2 to LATS1/2 (Lallemand et al., 2003, Genes Dev 17, 1090-1100; Yin et al., 2013, Dev Cell 19, 27-38). The LATS kinases will in turn phosphorylate and inactivate two highly homologous transcriptional co-activators: Yes-associated Protein (YAP) and Transcriptional co-activator with PDZ-binding motif (TAZ) by cytoplasmic sequestration via 14-3-3 and by ubiquitin-mediated degradation induced by β-TRCP E3 ligase. When the Hippo pathway is inactive, YAP and TAZ translocate in the nucleus to bind to the TEAD transcription factor family to induce expression of a specific signature promoting matrix remodeling, cell proliferation, survival and migration. TEAD1-4 can also bind to VGLL4 in the nucleus and act as a transcriptional repressor. VGLL4 is not structurally related to YAP/TAZ, but competes with YAP/TAZ based on a partially overlapping binding site on TEAD (Johnson and Halder, 2014, Nat Rev Drug Discov 13, 63-79).


TEADs are evolutionarily conserved proteins required for cardiogenesis, myogenesis, and for the development of the neural crest, notochord, and trophoectoderm. In mammals, there are four genes encoding four homologous members of the TEAD family named TEAD1-4. Each TEAD gene has a distinct but not mutually exclusive expression pattern. All TEAD family members are controlled by YAP/TAZ.


In fruit flies, loss of function of Hippo or Warts kinases (MST1/2 or LATS1/2 in mammals), or overexpression of Yorkie (the Drosophila homolog of YAP and TAZ), results in a dramatic overgrowth of the cuticle, as a result of dysregulated cell proliferation and resistance to apoptosis, leading to increased organ size. In mice, YAP overexpression, loss of MST1/2 or LATS1/2 kinase activities, or loss of NF2 leads to TEAD target gene up-regulation and progenitor cell expansion, resulting in liver and cardiac overgrowth and ultimately cancer formation in the liver, the small intestine and in skin. In contrast, a serine to alanine mutation at position 94 in YAP, that is unable to bind to TEAD, is not oncogenic (Zhao et al., 2008, Genes Dev 22, 1962-1971). Likewise, a dominant-negative TEAD mutant that is unable to bind DNA, overcomes YAP-driven liver tumorigenesis. In addition, NF2 mutant liver carcinoma was greatly suppressed by heterozogous loss of Yap (Zhang et al., 2010, Dev Cell 19, 27-38). Finally, verteporfin, a small molecule that inhibits YAP-TEAD association significantly suppressed the oncogenic activity of YAP in these models (Liu-Chittenden et al., 2012, Genes Dev 26, 1300-1305).


Gene amplification of YAP1 (encoding for YAP) and WWTR1 (encoding for TAZ) as well as constitutive nuclear localization of YAP/TAZ have been reported in many human solid malignancies, including liver, lung, breast, skin, colon and ovarian cancer and YAP/TAZ promote the acquisition of several important cancer cell phenotypes, such as proliferation, resistance to apoptosis, invasion, and immune-suppression (e.g. by attracting myeloid derived suppressor cells (Wang et al., 2016, Cancer Discov 6, 80-95)). In addition, gene fusions with YAP1 have been identified in several cancer types including ependymomas, vascular cancers, cervical carcinomas and porocarcinomas, which results in constitutive activation of YAP-TEAD, and are oncogenic in mice (Szulzewsky et al., 2020, Genes Dev 34: 1-14). In addition, several germline or somatic mutations in components of the Hippo pathway associated with various cancer types have been discovered in targeted and whole-genome sequencing studies. The best studied example is the NF2 locus, mutated with a high frequency in neurofibromatosis. Loss of NF2 and LATS2 are also frequently observed in schwannomas. Another tumor type that is commonly (in about 70% of all cases) associated with constitutive YAP-TEAD activation through genetic inactivation of NF2, LATS1/2, MST1/2 or SAV1, is malignant mesothelioma (Bueno et al., 2016, Nat Genet 48, 407-416.). Recent studies have shown that several mesothelioma cell lines with NF2 loss-of-function mutations exhibit a decrease in YAP phosphorylation and an increase in YAP-TEAD reporter activity. The YAP-TEAD transcription and viability of NF2 mutant mesothelioma cell lines (but not WT mesothelioma) are sensitive to YAP siRNA (an effect which can be rescued by overexpression of siRNA resistant YAP) and to treatment with verteporfin, a YAP antagonist (Zhang et al., 2017, J Cell Mol Med 21: 2663-2676).


Nuclear YAP has also emerged as a critical mediator of WNT dependent colorectal tumorigenesis. YAP-TEAD mediated transcription of genes involved in proliferation and stem cell renewal cooperate with WNT driven beta-catenin, and YAP is required for formation of adenomas following APC (adenomatous polyposis coli) inactivation (Azzolin et al., 2014 Cell 158, 157-170; Gregorieff et al., 2015 Nature 526, 715-718.). Recently, TIAM1, was identified as a suppressor of aggressive, metastatic colorectal cancer (CRC) by antagonizing YAP-TEAD transcription, again highlighting the role of YAP-TEAD in CRC (Diamantopoulou et al., 2017 Cancer Cell 31, 621-634).


In summary, YAP/TAZ activation has been shown to drive tumorigenesis and YAP/TAZ is hyperactivated in many different types of cancer in humans (often through loss-of-function mutations in upstream negative regulators). Genetic deletion or pharmacologic inhibition of YAP/TAZ has been shown to suppress tumor development and progression in different types of cancer. Therefore, it is believed that deregulation of the Hippo tumor suppressor pathway is a major event in the development of a wide range of cancer types and malignancies. Hence, pharmacological targeting of the Hippo cascade through inhibition of YAP, TAZ, TEAD, and/or the YAP/TAZ-TEAD protein-protein interaction would be a valuable approach for the treatment of cancers that harbor functional alterations of this pathway.


YAP/TAZ-TEAD activation has also been shown to play an important role in other diseases than cancer, namely such as in fibrosis and certain congenital disorders. A hallmark of fibrosis is the excessive deposition of extracellular matrix (ECM), including cross-linked collagen fibres, which results in the stiffening of tissues and eventually in dysfunctioning of affected organs. ECM stiffening promotes the nuclear activity of YAP/TAZ in cancer-associated fibroblasts, and fibroblasts of the liver, kidney, lung and skin (Mannaerts et al., 2015, J. Hepatol. 63, 679-688; Piersma et al., 2015, Am. J. Pathol. 185, 3326-3337). Nuclear YAP/TAZ promotes fibrotic cellular phenotypes, such as myofibroblast differentiation and increased matrix remodeling. Several genes that encode key secreted factors implicated in fibrosis are direct YAP/TAZ-TEAD targets. These genes include well-characterized pro-fibrotic factors, such as connective tissue growth factor (CTGF), plasminogen activator inhibitor 1 (PAI-1) and the lysyl oxidase (LOX) family of collagen cross-linking enzymes. Several lines of evidence support YAP/TAZ as contributors to fibrotic disease in vivo. These include reports of elevated YAP/TAZ levels and transcriptional activity in fibroblasts as well as in alveolar and respiratory epithelium of patients with idiopathic pulmonary fibrosis (Gokey et al., 2018 JCI Insight 3: e98738). Increased nuclear YAP has also been observed in patients with primary sclerosing cholangitis and primary biliary cirrhosis, which are chronic fibrotic disorders of liver injury. Expression of YAP or TAZ in the duct cells of the liver drives fibrosis progression that parallels fibrosis in nonalcoholic fatty liver disease (Machado et al., 2015, J. Hepatol 63, 962-970). Collectively, these studies suggest that targeting aberrant YAP/TAZ activity in fibrotic diseases may hold promise for therapy.


Neurofibromatosis type 2 is characterized by nervous system tumors including schwannomas, meningiomas, and ependymomas. Neurofibromatosis type 2 is an inheritable disorder caused by the inactivation of NF2 (Striedinger et al., 2008, Neoplasia 10, 1204-1210). Loss of NF2 leads to constitutive activation of YAP/TAZ-TEAD. The Sturge-Weber syndrome is a congenital eurocutaneous disorder characterized by a port-wine stain affecting the skin in the distribution of the ophthalmic branch of the trigeminal nerve, abnormal capillary venous vessels in the leptomeninges of the brain and choroid, glaucoma, seizures, stroke, and intellectual disability. The Sturge-Weber syndrome and port-wine stains are caused by a somatic activating mutation in GNAQ which leads to activation of YAP/TAZ-TEAD transcription (Shirley et al., 2013, NEJM, 368, 1971-1979). Therefore, several congenital disorders, characterized by constitutive YAP/TAZ-TEAD activation could be treated with inhibitors of YAP/TAZ-TEAD.


A few publications describe inhibitors of the YAP-TEAD transcriptional activation. Inventiva highlighted YAP-TEAD protein-protein interaction inhibitors in WO2020/070181, WO2018/185266, and WO2017/064277. The General Hospital Corporation, Boston described autopalmitoylation inhibitors in WO2017/053706. Vivace Therapeutics, Inc. disclosed non-fused tricyclic (WO2018/204532), benzosulfonyl (WO2019/040380), benzocarbonyl (WO2019/113236), oxadiazole (WO2019/222431), and bicyclic (WO2020/097389) compounds that modulate the interaction between YAP/TAZ and TEAD. The Regents of the University of California and Vivace Therapeutics, Inc. described tricyclic compounds that inhibit the Hippo-YAP signaling pathway in WO2013/188138 and WO2017/058716, respectively. Kyowa Hakko Kirin Co., Ltd. revealed alpha,beta-unsaturated amide compounds that display anti-cancer activity in WO2018/235926 and US2019/0010136. Genentech, Inc. disclosed carboxamide and sulfonamide derivatives useful as inhibitors of the YAP-TEAD protein-protein interaction in WO2019/232216 and WO2020/051099. Dana-Farber Cancer Institute, Inc. highlighted inhibitors of TEAD transcription factors in WO2020/081572. The Trustees of Indiana University described small-molecules that bind within the hydrophobic palmitate-binding pocket of TEADs in WO2020/087063. Wenchao Lu, et al. published vinylsulfonamides as covalent TEAD autopalmitoylation inhibitors (2019, European Journal of Medicinal Chemistry, 184, p. 111767). Korean Research Institute of Chemical Technology disclosed benzo[cd]indol-2(1H)-one derivatives that inhibit YAP-TEAD binding.


However, there is still a great need for novel, alternative or better therapeutics for the prevention or treatment of diseases mediated by the YAP/TAZ-TEAD activation, such as cancer and fibrosis among potentially other indications. Therapeutics with better potency, less side-effects, a higher activity, a lower toxicity or better pharmacokinetic or -dynamic properties or combinations thereof would be very welcome.


The present disclosure provides a class of novel compounds which can be used as inhibitors of the YAP/TAZ-TEAD activation mediated diseases.


SUMMARY OF THE DISCLOSURE

The present disclosure is based on the finding that at least one of the above-mentioned problems can be solved by the below described class of compounds.


The present disclosure provides new compounds which have been shown to possess inhibitory activity on the YAP/TAZ-TEAD transcription. The present disclosure furthermore demonstrates that these compounds efficiently inhibit the activity of YAP/TAZ-TEAD transcription. Therefore, these compounds constitute a useful class of new potent compounds that can be used in the treatment and/or prevention of Hippo mediated disorders in animals, mammals and humans, more specifically for the treatment and/or prevention of (i) cancer, more specifically lung cancer, breast cancer, head and neck cancer, oesophageal cancer, kidney cancer, bladder cancer, colon cancer, ovarian cancer, cervical cancer, endometrial cancer, liver cancer (including but not limited to cholangiocarcinoma), skin cancer, pancreatic cancer, gastric cancer, brain cancer and prostate cancer, mesotheliomas, and/or sarcomas (ii) fibrosis, and (iii) YAP/TAZ-TEAD activation related congenital disorders, among others.


In some aspects, the compounds described herein can be used in the treatment and/or prevention of Hippo mediated disorders in animals, mammals and humans, more specifically for the treatment and/or prevention of acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute T-cell leukemia, basal cell carcinoma, bile duct carcinoma, bronchogenic carcinoma, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, glioblastoma, gliosarcoma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, leukemia, liposarcoma, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, NUT midline carcinoma (NMC), non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenstrom's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor.


The present disclosure furthermore relates for the use of such compounds as medicines and to their use for the manufacture of medicaments, more in particular for treating and/or preventing YAP/TAZ-TEAD activation mediated diseases, in particular (i) cancer, more specifically lung cancer, breast cancer, head and neck cancer, oesophageal cancer, kidney cancer, bladder cancer, colon cancer, ovarian cancer, cervical cancer, endometrial cancer, liver cancer (including but not limited to cholangiocarcinoma), skin cancer, pancreatic cancer, gastric cancer, brain cancer and prostate cancer, mesotheliomas, and/or sarcomas and (ii) fibrosis in animals or mammals, more in particular in humans. The disclosure also relates to methods for the preparation of all such compounds and to pharmaceutical compositions comprising them in an effective amount.


In some embodiments, the disclosure relates to the compounds of the invention for use as a medicine, to the use of such compounds as medicines and to their use for the manufacture of medicaments, more in particular for treating and/or preventing YAP/TAZ-TEAD activation mediated diseases more specifically for the treatment and/or prevention of acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute T-cell leukemia, basal cell carcinoma, bile duct carcinoma, bronchogenic carcinoma, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, glioblastoma, gliosarcoma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, leukemia, liposarcoma, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, NUT midline carcinoma (NMC), non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenstrom's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor.


The present disclosure also relates to a method of treatment or prevention of TEAD activation mediated disorders in humans by the administration of one or more such compounds, optionally in combination with one or more other medicines, to a patient in need thereof. The present disclosure also relates to methods of preparing the compounds disclosed herein comprising the steps for synthesis of the compounds described herein.







DETAILED DESCRIPTION
Definitions

The term “YAP/TAZ-TEAD activation mediated diseases” refers to diseases in which hippo signaling is inactivated and whereby YAP/TAZ-TEAD activation is contributing, driving, sustaining, enabling or the like such disease. This might be through loss-of-function mutations or deletions in the genes encoding the upstream regulators of YAP/TAZ-TEAD (e.g. NF2, MST1/2, LATS1/2, FAT1 or SAV1), unleashing constitutive YAP-TEAD transcriptional activity leading to unbridled tumor growth and metastasis of some cancers. This might also be through YAP1 or WWTR1 (TAZ) gene amplifications, gene fusions or activating mutations, or YAP/TAZ overexpression or hyperactivity, among others. YAP/TAZ-TEAD activation mediated diseases therefore refers to cancer, but also includes fibrosis and certain congential disorders. Cancers that are included in YAP/TAZ-TEAD mediated diseases are, without being limited thereto, lung cancer, breast cancer, head and neck cancer, oesophageal cancer, kidney cancer, bladder cancer, colon cancer, ovarian cancer, cervical cancer, endometrial cancer, liver cancer (including but not limited to cholangiocarcinoma), skin cancer, pancreatic cancer, gastric cancer, brain cancer and prostate cancer, mesotheliomas, and/or sarcomas. Also included are (i) squamous cell carcinomas of the lung, cervix, ovaries, head and neck, oesophagus, and/or skin, or (ii) cancers that originate from neuroectoderm-derived tissues, such as ependymomas, meningiomas, schwannomas, peripheral nerve-sheet tumors and/or neuroblastomas, or (iii) vascular cancers, such as epithelioid haemangioendotheliomas. Fibrotic diseases or fibrosis that is included in YAP/TAZ-TEAD mediated diseases are, without being limited thereto, liver fibrosis, lung fibrosis and heart fibrosis. Congenital disorders that are included in YAP/TAZ-TEAD mediated diseases are, without being limited thereto, Sturge-Weber syndrome and Neurofibromatosis type 2.


YAP/TAZ-TEAD mediated diseases also includes cancers that have developed resistance to prior treatments such has EGFR inhibitors, MEK inhibitors, AXL inhibitors, B-RAF inhibitors, RAS inhibitors and others.


The term “treat” or “treating” as used herein is intended to refer to administration of a compound or composition to a subject for the purpose of effecting a therapeutic benefit or prophylactic benefit through inhibition of the YAP/TAZ-TEAD transcription. Treating includes reversing, ameliorating, alleviating, inhibiting the progress of, lessening the severity of, or preventing a disease, disorder, or condition, or one or more symptoms of such disease, disorder or condition mediated through YAP/TAZ-TEAD transcription. By “therapeutic benefit” is meant eradication, amelioration, reversing, alleviating, inhibiting the progress of or lessening the severity of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient is afflicted with the underlying disorder in some embodiments. For prophylactic benefit, in some embodiments, the compositions are administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease has not been made.


The term “subject” as used herein, refers to an animal, for example a mammal, such as a human, a patient, who has been the object of treatment, observation or experiment or who is in need of such treatment.


The term “therapeutically effective amount” as used herein, means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation or partial alleviation of the symptoms of the disease or disorder being treated.


The term “composition” as used herein is intended to encompass a product comprising the specified ingredients in the therapeutically effective amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts.


The term “antagonist” or “inhibitor” as used herein in reference to inhibitors of the YAP/TAZ-TEAD activation, refers to a compound capable of producing, depending on the circumstance, a functional antagonism of YAP/TAZ-TEAD activation.


It is to be noticed that the term “comprising”, used in the claims, should not be interpreted as being restricted to the means listed thereafter; it does not exclude other elements or steps.


Reference throughout this specification to “one embodiment” or “an embodiment” means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Furthermore, the particular features, structures or characteristics may be combined in any suitable manner, as would be apparent to one of ordinary skill in the art from this disclosure, in one or more embodiments. Where an indefinite or definite article is used when referring to a singular noun e.g. “a” or “an”, “the”, this includes a plural of that noun unless something else is specifically stated.


Similarly it should be appreciated that in the description of exemplary embodiments of the disclosure, various features of the disclosure are sometimes grouped together in a single embodiment, figure, or description thereof for the purpose of streamlining the disclosure and aiding in the understanding of one or more of the various inventive aspects.


In each of the following definitions, the number of carbon atoms represents the maximum number of carbon atoms generally optimally present in the substituent or linker; it is understood that where otherwise indicated in the present application, the number of carbon atoms represents the optimal maximum number of carbon atoms for that particular substituent or linker.


The term “leaving group” or “LG” as used herein means a chemical group which is susceptible to be displaced by a nucleophile or cleaved off or hydrolyzed in basic or acidic conditions. In a particular embodiment, a leaving group is selected from a halogen atom (e.g., Cl, Br, I) or a sulfonate (e.g., mesylate, tosylate, triflate).


The term “protecting group” refers to a moiety of a compound that masks or alters the properties of a functional group or the properties of the compound as a whole. The chemical substructure of a protecting group varies widely. One function of a protecting group is to serve as intermediates in the synthesis of the parental drug substance. Chemical protecting groups and strategies for protection/deprotection are well known in the art. See: “Protective Groups in Organic Chemistry”, Theodora W. Greene (John Wiley & Sons, Inc., New York, 1991. Protecting groups are often utilized to mask the reactivity of certain functional groups, to assist in the efficiency of desired chemical reactions, e.g. making and breaking chemical bonds in an ordered and planned fashion. Protection of functional groups of a compound alters other physical properties besides the reactivity of the protected functional group, such as the polarity, lipophilicity (hydrophobicity), and other properties which can be measured by common analytical tools. Chemically protected intermediates may themselves be biologically active or inactive.


Protected compounds may also exhibit altered, and in some cases, optimized properties in vitro and in vivo, such as passage through cellular membranes and resistance to enzymatic degradation or sequestration. In this role, protected compounds with intended therapeutic effects may be referred to as prodrugs. Another function of a protecting group is to convert the parental drug into a prodrug, whereby the parental drug is released upon conversion of the prodrug in vivo. Because active prodrugs may be absorbed more effectively than the parental drug, prodrugs may possess greater potency in vivo than the parental drug. Protecting groups are removed either in vitro, in the instance of chemical intermediates, or in vivo, in the case of prodrugs. With chemical intermediates, it is not particularly important that the resulting products after deprotection, e.g. alcohols, be physiologically acceptable, although in general it is more desirable if the products are pharmacologically innocuous.


The term “alkyl” or “C1-18alkyl” as used herein means C1-C18 normal, secondary, or tertiary, linear, branched or straight hydrocarbon with no site of unsaturation. Examples are methyl, ethyl, 1-propyl (n-propyl), 2-propyl (iPr), 1-butyl, 2-methyl-1-propyl(i-Bu), 2-butyl (s-Bu), 2-dimethyl propyl (t-Bu), 1-pentyl (n-pentyl), 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3-dimethyl-2-butyl, 3,3-dimethyl-2-butyl, n-heptyl, n-octyl, n-nonyl, n-decyl, n-undecyl, n-dodecyl, n-tridecyl, n-tetradecyl, n-pentadecyl, n-hexadecyl, n-heptadecyl, n-octadecyl, n-nonadecyl, and n-icosyl. In particular embodiments, the term alkyl refers to C1-12alkyl (C1-12 hydrocarbons), yet more in particular to C1-9alkyl (C1-9 hydrocarbons), yet more in particular to C1-6alkyl (C1-6 hydrocarbons) as further defined herein above.


The term “haloalkyl” as a group or part of a group, refers to an alkyl group having the meaning as defined above wherein one, two, or three hydrogen atoms are each replaced with a halogen as defined herein. Non-limiting examples of such haloalkyl groups include chloromethyl, 1-bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1,1,1-trifluoroethyl and the like.


The term “alkoxy” or “alkyloxy”, as a group or part of a group, refers to a group having the formula —ORb wherein Rb is C1-6alkyl as defined herein above. Non-limiting examples of suitable C1-6alkoxy include methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentyloxy and hexyloxy.


The term “haloalkoxy”, as a group or part of a group, refers to a group of formula —O—Rc, wherein Rc is haloalkyl as defined herein. Non-limiting examples of suitable haloalkoxy include fluoromethoxy, difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, 1,1,2,2-tetrafluoroethoxy, 2-fluoroethoxy, 2-chloroethoxy, 2,2-difluoroethoxy, 2,2,2-trichloroethoxy, trichloromethoxy, 2-bromoethoxy, pentafluoroethyl, 3,3,3-trichloropropoxy, 4,4,4-trichlorobutoxy.


The term “cycloalkyl” or “C3-18 cycloalkyl” as used herein and unless otherwise stated means a saturated hydrocarbon monovalent group having from 3 to 18 carbon atoms consisting of or comprising a C3-10 monocyclic or C7-18 polycyclic saturated hydrocarbon, such as for instance cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylethylene, methylcyclopropylene, cyclohexyl, cycloheptyl, cyclooctyl, cyclooctylmethylene, norbornyl, fenchyl, trimethyltricycloheptyl, decalinyl, adamantyl and the like. In particular embodiments, the term cycloalkyl refers to C3-12 cycloalkyl (saturated cyclic C3-12hydrocarbons), yet more in particular to C3-9cycloalkyl (saturated cyclic C3-9hydrocarbons), still more in particular to C3-6cycloalkyl (saturated cyclic C3-6hydrocarbons) as further defined herein above. For the avoidance of doubt, fused systems of a cycloalkyl ring with a heterocyclic ring are considered as heterocycle irrespective of the ring that is bound to the core structure. Fused systems of a cycloalkyl ring with an aryl ring are considered as aryl irrespective of the ring that is bound to the core structure. Fused systems of a cycloalkyl ring with a heteroaryl ring are considered as heteroaryl irrespective of the ring that is bound to the core structure.


The term “alkenyl” or “C2-18alkenyl” as used herein is C2-C18 normal, secondary or tertiary, linear, branched or straight hydrocarbon with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp2 double bond. Examples include, but are not limited to: ethylene or vinyl (—CH═CH2), allyl (—CH2CH═CH2), and 5-hexenyl (—CH2CH2CH2CH2CH═CH2). The double bond may be in the cis or trans configuration. In particular embodiments, the term alkenyl refers to C2-12alkenyl (C2-12hydrocarbons), yet more in particular to C2-9 alkenyl (C2-9 hydrocarbons), still more in particular to C2-6 alkenyl (C2-6hydrocarbons) as further defined herein above with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp2 double bond.


The term “alkenyloxy”, as a group or part of a group, refers to a group having the formula —ORd wherein Rd is alkenyl as defined herein above.


The term “cycloalkenyl” as used herein refers to a non-aromatic hydrocarbon group having from 5 to 18 carbon atoms with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp2 double bond and consisting of or comprising a C6-10 monocyclic or C7-18 polycyclic hydrocarbon. Examples include, but are not limited to: cyclopentenyl (—C6H7), cyclopentenylpropylene, methylcyclohexenylene and cyclohexenyl (—C6H9). The double bond may be in the cis or trans configuration. In particular embodiments, the term cycloalkenyl refers to C5-12 cycloalkenyl (cyclic C5-12 hydrocarbons), yet more in particular to C5-9 cycloalkenyl (cyclic C5-9 hydrocarbons), still more in particular to C5-6 cycloalkenyl (cyclic C5-6 hydrocarbons) as further defined herein above with at least one site of unsaturation, namely a carbon-carbon, sp2 double bond. For the avoidance of doubt, fused systems of a cycloalkenyl ring with a heterocyclic ring are considered as heterocycle irrespective of the ring that is bound to the core structure. Fused systems of a cycloalkenyl ring with an aryl ring are considered as aryl irrespective of the ring that is bound to the core structure. Fused systems of a cycloalkenyl ring with a heteroaryl ring are considered as heteroaryl irrespective of the ring that is bound to the core structure.


The term “alkynyl” or “C2-18alkynyl” as used herein refers to C2-C18 normal, secondary, tertiary, linear, branched or straight hydrocarbon with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp triple bond. Examples include, but are not limited to: ethynyl (—C≡CH), 3-ethyl-cyclohept-1-ynylene, and 1-propynyl (propargyl, —CH2C≡CH). In particular embodiments, the term alkynyl refers to C2-12 alkynyl (C2-12 hydrocarbons), yet more in particular to C2-9 alkynyl (C2-9 hydrocarbons) yet more in particular to C2-6 alkynyl (C2-6 hydrocarbons) as further defined herein above with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp triple bond.


The term “alkynyloxy”, as a group or part of a group, refers to a group having the formula —ORe wherein Re is alkynyl as defined herein above.


The term “cycloalkynyl” as used herein refers to a non-aromatic hydrocarbon group having from 5 to 18 carbon atoms with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp triple bond and consisting of or comprising a C5-10 monocyclic or C7-18 polycyclic hydrocarbon. Examples include, but are not limited to: cyclohept-1-yne, 3-ethyl-cyclohept-1-ynylene, 4-cyclohept-1-yn-methylene and ethylene-cyclohept-1-yne. In particular embodiments, the term cycloalkynyl refers to C5-10 cycloalkynyl (cyclic C6-10 hydrocarbons), yet more in particular to C5-9 cycloalkynyl (cyclic C5-9 hydrocarbons), still more in particular to C6-6 cycloalkynyl (cyclic C5-6 hydrocarbons) as further defined herein above with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp triple bond. For the avoidance of doubt, fused systems of a cycloalkynyl ring with a heterocyclic ring are considered as heterocycle irrespective of the ring that is bound to the core structure. Fused systems of a cycloalkynyl ring with an aryl ring are considered as aryl irrespective of the ring that is bound to the core structure. Fused systems of a cycloalkynyl ring with a heteroaryl ring are considered as heteroaryl irrespective of the ring that is bound to the core structure.


The term “alkylene” as used herein each refer to a saturated, branched or straight chain hydrocarbon group of 1-18 carbon atoms (more in particular C1-12, C1-9 Or C1-6 carbon atoms), and having two monovalent group centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Typical alkylene include, but are not limited to: methylene (—CH2—), 1,2-ethyl (—CH2CH2—), 1,3-propyl (—CH2CH2CH2—), 1,4-butyl (—CH2CH2CH2CH2—), and the like.


The term “alkenylene” as used herein each refer to a branched or straight chain hydrocarbon of 2-18 carbon atoms (more in particular C2-12, C2-9 or C2-6 carbon atoms) with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp2 double bond, and having two monovalent centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene.


The term “alkynylene” as used herein each refer to a branched or straight chain hydrocarbon of 2-18 carbon atoms (more in particular C2-12, C2-9 or C2-6 carbon atoms) with at least one site (usually 1 to 3, preferably 1) of unsaturation, namely a carbon-carbon, sp triple bond, and having two monovalent centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne.


The term “heteroalkyl” as used herein refers to an alkyl wherein one or more carbon atoms are replaced by one or more atoms selected from the group comprising oxygen, nitrogen or sulphur atom. The term heteroalkyl thus comprises —O—Rb, —NRo—Rb, —Ra—O—Rb, and —S—Rb, wherein Ra is alkylene, Rb is alkyl, and Ro is hydrogen or alky as defined herein. In particular embodiments, the term refers to C1-12heteroalkyl, C1-9heteroalkyl or C1-6heteroalkyl. In some embodiments heteroalkyl is selected from the group comprising alkyloxy, alkyl-oxy-alkyl, (mono or di)alkylamino, (mono or di-)alkyl-amino-alkyl, alkylthio, and alkyl-thio-alkyl.


The term “heteroalkenyl” as used herein refers to an acyclic alkenyl wherein one or more carbon atoms are replaced by one or more atoms selected from oxygen, nitrogen or sulphur atom. The term heteroalkenyl thus comprises —O—Rd, —NH—(Rd), —N(Rd))2, —N(Rb)(Rd), and —S—Rd wherein Rb is alkyl and Rd is alkenyl as defined herein. In particular embodiments, the term refers to C2-12heteroalkenyl, C2-9heteroalkenyl or C2-6heteroalkenyl. In some embodiments heteroalkenyl is selected from the group comprising alkenyloxy, alkenyl-oxy-alkenyl, (mono or di-)alkenylamino, (mono or di-)alkenyl-amino-alkenyl, alkenylthio, and alkenyl-thio-alkenyl,


The term “heteroalkynyl” as used herein refers to an acyclic alkynyl wherein one or more carbon atoms are replaced by an oxygen, nitrogen or sulphur atom. The term heteroalkynyl thus comprises but is not limited to —O—Rd, —N(Rd)2, NHRd, —N(Rb)(Re), —N(Rd)(Re), and —S—Rd wherein Rb is alkyl, Re is alkynyl and Rd is alkenyl as defined herein. In particular embodiments, the term refers to C2-12heteroalkynyl, C2-9heteroalkynyl or C2-6heteroalkynyl. In some embodiments the term heteroalkynyl is selected from the group comprising alkynyloxy, alkynyl-oxy-alkynyl, (mono or di-)alkynylamino, (mono or di-)alkynyl-amino-alkynyl, alkynylthio, alkynyl-thio-alkynyl,


The term “heteroalkylene” as used herein refers to an alkylene wherein one or more carbon atoms are replaced by one or more oxygen, nitrogen or sulphur atoms.


The term “heteroalkenylene” as used herein refers to an alkenylene wherein one or more carbon atoms are replaced by one or more oxygen, nitrogen or sulphur atoms.


The term “heteroalkynylene” as used herein refers to an alkynylene wherein one or more carbon atoms are replaced by one or more oxygen, nitrogen or sulphur atom.


The term “aryl” as used herein means an aromatic hydrocarbon of 6-20 carbon atoms derived by the removal of hydrogen from a carbon atom of a parent aromatic ring system. Typical aryl groups include, but are not limited to 1 ring, or 2 or 3 rings fused together, derived from benzene, naphthalene, anthracene, biphenyl, and the like. In particular embodiments, the term aryl refers to a 6-14 carbon atoms membered aromatic cycle, yet more in particular refers to a 6-10 carbon atoms membered aromatic cycle. Fused systems of an aryl ring with a cycloalkyl ring, or a cycloalkenyl ring, or a cycloalkynyl ring, are considered as aryl irrespective of the ring that is bound to the core structure. Fused systems of an aryl ring with a heterocycle are considered as heterocycle irrespective of the ring that is bound to the core structure. Thus, indoline, dihydrobenzofurane, dihydrobenzothiophene and the like are considered as heterocycle according to the disclosure. Fused systems of an aryl ring with a heteroaryl ring are considered as heteroaryl irrespective of the ring that is bound to the core structure.


The term “aryloxy”, as a group or part of a group, refers to a group having the formula —ORg wherein Rg is aryl as defined herein above.


The term “arylalkyl” or “arylalkyl-” as used herein refers to an alkyl in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethyl, and the like. The arylalkyl group comprises 6 to 20 carbon atoms, e.g. the alkyl moiety of the arylalkyl group is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.


The term “arylalkyloxy”, as a group or part of a group, refers to a group having the formula —O—Ra—Rg wherein Rg is aryl, and Ra is alkylene as defined herein above.


The term “arylalkenyl” or “arylalkenyl-” as used herein refers to an alkenyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an aryl. The arylalkenyl group comprises 6 to 20 carbon atoms, e.g. the alkenyl moiety of the arylalkenyl group is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.


The term “arylalkynyl” or “arylalkynyl-” as used herein refers to an alkynyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an aryl. The arylalkynyl group comprises 6 to 20 carbon atoms, e.g. the alkynyl moiety of the arylalkynyl group is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.


The term “arylheteroalkyl” or “arylheteroalkyl-” as used herein refers to a heteroalkyl in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl. The arylheteroalkyl group comprises 6 to 20 carbon atoms, e.g. the heteroalkyl moiety of the arylheteroalkyl group is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. In some embodiments arylheteroalkyl is selected from the group comprising aryl-O-alkyl, arylalkyl-O-alkyl, aryl-NH-alkyl, aryl-N(alkyl)2, arylalkyl-NH-alkyl, arylalkyl-N-(alkyl)2, aryl-S-alkyl, and arylalkyl-S-alkyl.


The term “arylheteroalkenyl” or “arylheteroalkenyl-” as used herein refers to a heteroalkenyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an aryl. The arylheteroalkenyl group comprises 6 to 20 carbon atoms, e.g. the heteroalkenyl moiety of the arylheteroalkenyl group is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. In some embodiments arylheteroalkenyl is selected from the group comprising aryl-O-alkenyl, arylalkenyl-O-alkenyl, aryl-NH-alkenyl, aryl-N(alkenyl)2, arylalkenyl-NH-alkenyl, arylalkenyl-N-(alkenyl)2, aryl-S-alkenyl, and arylalkenyl-S-alkenyl.


The term “arylheteroalkynyl” or “arylheteroalkynyl-” as used herein refers to a heteroalkynyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an aryl. The arylheteroalkynyl group comprises 6 to 20 carbon atoms, e.g. the heteroalkynyl moiety of the arylheteroalkynyl group is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. In some embodiments arylheteroalkynyl is selected from the group comprising aryl-O-alkynyl, arylalkynyl-O-alkynyl, aryl-NH-alkynyl, aryl-N(alkynyl)2, arylalkynyl-NH-alkynyl, arylalkynyl-N-(alkynyl)2, aryl-S-alkynyl, and arylalkynyl-S-alkynyl.


The term “heterocycle” or “heterocyclyl” as used herein refer to non-aromatic, fully saturated or partially unsaturated ring system of 3 to 18 atoms including at least one N, O, S, or P (for example, 3 to 7 member monocyclic, 7 to 11 member bicyclic, or comprising a total of 3 to 10 ring atoms). Each ring of the heterocycle or heterocyclyl may have 1, 2, 3 or 4 heteroatoms selected from N, O and/or S, where the N and S heteroatoms may optionally be oxidized and the N heteroatoms may optionally be quaternized; and wherein at least one carbon atom of heterocyclyl can be oxidized to form at least one C═O. The heterocycle may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows. The rings of multi-ring heterocyclyls or heterocycles may be fused, bridged and/or joined through one or more spiro atoms. Fused systems of a heterocycle or heterocyclyl with an aryl ring are considered as heterocycle or heterocyclyl irrespective of the ring that is bound to the core structure. Fused systems of a heterocycle or heterocyclyl with a heteroaryl ring are considered as heteroaryl irrespective of the ring that is bound to the core structure.


Non limiting exemplary heterocycles or heterocyclic groups include piperidinyl, piperazinyl, homopiperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, aziridinyl, oxiranyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 2-imidazolinyl, pyrazolidinyl imidazolidinyl, isoxazolinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, succinimidyl, 3H-indolyl, indolinyl, isoindolinyl, chromanyl (also known as 3,4-dihydrobenzo[b]pyranyl), 2H-pyrrolyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, 4H-quinolizinyl, 2-oxopiperazinyl, 2-pyrazolinyl, 3-pyrazolinyl, tetrahydro-2H-pyranyl, 2H-pyranyl, 4H-pyranyl, 3,4-dihydro-2H-pyranyl, 3-dioxolanyl, 1,4-dioxanyl, 2,5-dioximidazolidinyl, 2-oxopiperidinyl, 2-oxopyrrolodinyl, indolinyl, tetrahydrothiophenyl, tetrahydroquinolinyl, tetrahydroisoquinolin-1-yl, tetrahydroisoquinolin-2-yl, tetrahydroisoquinolin-3-yl, tetrahydroisoquinolin-4-yl, thiomorpholin-4-yl, thiomorpholin ylsulfoxide, thiomorpholin-4-ylsulfone, 1,3-dioxolanyl, 1,4-oxathianyl, 1,4-dithianyl, 1,3,5-trioxanyl, 1H-pyrrolizinyl, tetrahydro-1,1-dioxothiophenyl, N-formylpiperazinyl, and morpholin yl. The term “aziridinyl” as used herein includes aziridin-1-yl and aziridin-2-yl. The term “oxyranyl” as used herein includes oxyranyl-2-yl. The term “thiiranyl” as used herein includes thiiran-2-yl. The term “azetidinyl” as used herein includes azetidin-1-yl, azetidin-2-yl and azetidin-3-yl. The term “oxetanyl” as used herein includes oxetan-2-yl and oxetan-3-yl. The term “thietanyl” as used herein includes thietan-2-yl and thietan-3-yl. The term “pyrrolidinyl” as used herein includes pyrrolidin-1-yl, pyrrolidin-2-yl and pyrrolidin-3-yl. The term “tetrahydrofuranyl” as used herein includes tetrahydrofuran-2-yl and tetrahydrofuran-3-yl. The term “tetrahydrothiophenyl” as used herein includes tetrahydrothiophen-2-yl and tetrahydrothiophen-3-yl. The term “succinimidyl” as used herein includes succinimid-1-yl and succininmid-3-yl. The term “dihydropyrrolyl” as used herein includes 2,3-dihydropyrrol-1-yl, 2,3-dihydro-1H-pyrrol-2-yl, 2,3-dihydro-1H-pyrrol-3-yl, 2,5-dihydropyrrol-1-yl, 2,5-dihydro-1H-pyrrol-3-yl and 2,5-dihydropyrrol-5-yl. The term “2H-pyrrolyl” as used herein includes 2H-pyrrol-2-yl, 2H-pyrrol-3-yl, 2H-pyrrol-4-yl and 2H-pyrrol-5-yl. The term “3H-pyrrolyl” as used herein includes 3H-pyrrol-2-yl, 3H-pyrrol-3-yl, 3H-pyrrol-4-yl and 3H-pyrrol-5-yl. The term “dihydrofuranyl” as used herein includes 2,3-dihydrofuran-2-yl, 2,3-dihydrofuran-3-yl, 2,3-dihydrofuran-4-yl, 2,3-dihydrofuran-5-yl, 2,5-dihydrofuran-2-yl, 2,5-dihydrofuran-3-yl, 2,5-dihydrofuran-4-yl and 2,5-dihydrofuran-5-yl. The term “dihydrothiophenyl” as used herein includes 2,3-dihydrothiophen-2-yl, 2,3-dihydrothiophen-3-yl, 2,3-dihydrothiophen-4-yl, 2,3-dihydrothiophen-5-yl, 2,5-dihydrothiophen-2-yl, 2,5-dihydrothiophen-3-yl, 2,5-dihydrothiophen-4-yl and 2,5-dihydrothiophen-5-yl. The term “imidazolidinyl” as used herein includes imidazolidin-1-yl, imidazolidin-2-yl and imidazolidin-4-yl. The term “pyrazolidinyl” as used herein includes pyrazolidin-1-yl, pyrazolidin-3-yl and pyrazolidin-4-yl. The term “imidazolinyl” as used herein includes imidazolin-1-yl, imidazolin-2-yl, imidazolin-4-yl and imidazolin-5-yl. The term “pyrazolinyl” as used herein includes 1-pyrazolin-3-yl, 1-pyrazolin-4-yl, 2-pyrazolin-1-yl, 2-pyrazolin-3-yl, 2-pyrazolin-4-yl, 2-pyrazolin-5-yl, 3-pyrazolin-1-yl, 3-pyrazolin-2-yl, 3-pyrazolin-3-yl, 3-pyrazolin-4-yl and 3-pyrazolin-5-yl. The term “dioxolanyl” also known as “1,3-dioxolanyl” as used herein includes dioxolan-2-yl, dioxolan-4-yl and dioxolan-5-yl. The term “dioxolyl” also known as “1,3-dioxolyl” as used herein includes dioxol-2-yl, dioxol-4-yl and dioxol-5-yl. The term “oxazolidinyl” as used herein includes oxazolidin-2-yl, oxazolidin-3-yl, oxazolidin-4-yl and oxazolidin-5-yl. The term “isoxazolidinyl” as used herein includes isoxazolidin-2-yl, isoxazolidin-3-yl, isoxazolidin-4-yl and isoxazolidin-5-yl. The term “oxazolinyl” as used herein includes 2-oxazolinyl-2-yl, 2-oxazolinyl-4-yl, 2-oxazolinyl-5-yl, 3-oxazolinyl-2-yl, 3-oxazolinyl-4-yl, 3-oxazolinyl-5-yl, 4-oxazolinyl-2-yl, 4-oxazolinyl-3-yl, 4-oxazolinyl-4-yl and 4-oxazolinyl-5-yl. The term “isoxazolinyl” as used herein includes 2-isoxazolinyl-3-yl, 2-isoxazolinyl-4-yl, 2-isoxazolinyl-5-yl, 3-isoxazolinyl-3-yl, 3-isoxazolinyl-4-yl, 3-isoxazolinyl-5-yl, 4-isoxazolinyl-2-yl, 4-isoxazolinyl-3-yl, 4-isoxazolinyl-4-yl and 4-isoxazolinyl-5-yl. The term “thiazolidinyl” as used herein includes thiazolidin-2-yl, thiazolidin-3-yl, thiazolidin-4-yl and thiazolidin-5-yl. The term “isothiazolidinyl” as used herein includes isothiazolidin-2-yl, isothiazolidin-3-yl, isothiazolidin-4-yl and isothiazolidin-5-yl. The term “thiazolinyl” as used herein includes 2-thiazolinyl-2-yl, 2-thiazolinyl-4-yl, 2-thiazolinyl-5-yl, 3-thiazolinyl-2-yl, 3-thiazolinyl-4-yl, 3-thiazolinyl-5-yl, 4-thiazolinyl-2-yl, 4-thiazolinyl-3-yl, 4-thiazolinyl-4-yl and 4-thiazolinyl-5-yl. The term “isothiazolinyl” as used herein includes 2-isothiazolinyl-3-yl, 2-isothiazolinyl-4-yl, 2-isothiazolinyl-5-yl, 3-isothiazolinyl-3-yl, 3-isothiazolinyl-4-yl, 3-isothiazolinyl-5-yl, 4-isothiazolinyl-2-yl, 4-isothiazolinyl-3-yl, 4-isothiazolinyl-4-yl and 4-isothiazolinyl-5-yl. The term “piperidyl” also known as “piperidinyl” as used herein includes piperid-1-yl, piperid-2-yl, piperid-3-yl and piperid-4-yl. The term “dihydropyridinyl” as used herein includes 1,2-dihydropyridin-1-yl, 1,2-dihydropyridin-2-yl, 1,2-dihydropyridin-3-yl, 1,2-dihydropyridin-4-yl, 1,2-dihydropyridin-5-yl, 1,2-dihydropyridin-6-yl, 1,4-dihydropyridin-1-yl, 1,4-dihydropyridin-2-yl, 1,4-dihydropyridin-3-yl, 1,4-dihydropyridin-4-yl, 2,3-dihydropyridin-2-yl, 2,3-dihydropyridin-3-yl, 2,3-dihydropyridin-4-yl, 2,3-dihydropyridin-5-yl, 2,3-dihydropyridin-6-yl, 2,5-dihydropyridin-2-yl, 2,5-dihydropyridin-3-yl, 2,5-dihydropyridin-4-yl, 2,5-dihydropyridin-5-yl, 2,5-dihydropyridin-6-yl, 3,4-dihydropyridin-2-yl, 3,4-dihydropyridin-3-yl, 3,4-dihydropyridin-4-yl, 3,4-dihydropyridin-5-yl and 3,4-dihydropyridin-6-yl. The term “tetrahydropyridinyl” as used herein includes 1,2,3,4-tetrahydropyridin-1-yl, 1,2,3,4-tetrahydropyridin-2-yl, 1,2,3,4-tetrahydropyridin-3-yl, 1,2,3,4-tetrahydropyridin-4-yl, 1,2,3,4-tetrahydropyridin-5-yl, 1,2,3,4-tetrahydropyridin-6-yl, 1,2,3,6-tetrahydropyridin-1-yl, 1,2,3,6-tetrahydropyridin-2-yl, 1,2,3,6-tetrahydropyridin-3-yl, 1,2,3,6-tetrahydropyridin-4-yl, 1,2,3,6-tetrahydropyridin-5-yl, 1,2,3,6-tetrahydropyridin-6-yl, 2,3,4,5-tetrahydropyridin-2-yl, 2,3,4,5-tetrahydropyridin-3-yl, 2,3,4,5-tetrahydropyridin-3-yl, 2,3,4,5-tetrahydropyridin-4-yl, 2,3,4,5-tetrahydropyridin-5-yl and 2,3,4,5-tetrahydropyridin-6-yl. The term “tetrahydropyranyl” also known as “oxanyl” or “tetrahydro-2H-pyranyl”, as used herein includes tetrahydropyran-2-yl, tetrahydropyran-3-yl and tetrahydropyran-4-yl. The term “2H-pyranyl” as used herein includes 2H-pyran-2-yl, 2H-pyran-3-yl, 2H-pyran-4-yl, 2H-pyran-5-yl and 2H-pyran-6-yl. The term “4H-pyranyl” as used herein includes 4H-pyran-2-yl, 4H-pyran-3-yl and 4H-pyran-4-yl. The term “3,4-dihydro-2H-pyranyl” as used herein includes 3,4-dihydro-2H-pyran-2-yl, 3,4-dihydro-2H-pyran-3-yl, 3,4-dihydro-2H-pyran-4-yl, 3,4-dihydro-2H-pyran-5-yl and 3,4-dihydro-2H-pyran-6-yl. The term “3,6-dihydro-2H-pyranyl” as used herein includes 3,6-dihydro-2H-pyran-2-yl, 3,6-dihydro-2H-pyran-3-yl, 3,6-dihydro-2H-pyran-4-yl, 3,6-dihydro-2H-pyran-5-yl and 3,6-dihydro-2H-pyran-6-yl. The term “tetrahydrothiophenyl”, as used herein includes tetrahydrothiophen-2-yl, tetrahydrothiophenyl-3-yl and tetrahydrothiophenyl-4-yl. The term “2H-thiopyranyl” as used herein includes 2H-thiopyran-2-yl, 2H-thiopyran-3-yl, 2H-thiopyran-4-yl, 2H-thiopyran-5-yl and 2H-thiopyran-6-yl. The term “4H-thiopyranyl” as used herein includes 4H-thiopyran-2-yl, 4H-thiopyran-3-yl and 4H-thiopyran-4-yl. The term “3,4-dihydro-2H-thiopyranyl” as used herein includes 3,4-dihydro-2H-thiopyran-2-yl, 3,4-dihydro-2H-thiopyran-3-yl, 3,4-dihydro-2H-thiopyran-4-yl, 3,4-dihydro-2H-thiopyran-5-yl and 3,4-dihydro-2H-thiopyran-6-yl. The term “3,6-dihydro-2H-thiopyranyl” as used herein includes 3,6-dihydro-2H-thiopyran-2-yl, 3,6-dihydro-2H-thiopyran-3-yl, 3,6-dihydro-2H-thiopyran-4-yl, 3,6-dihydro-2H-thiopyran-5-yl and 3,6-dihydro-2H-thiopyran-6-yl. The term “piperazinyl” also known as “piperazidinyl” as used herein includes piperazin-1-yl and piperazin-2-yl. The term “morpholinyl” as used herein includes morpholin-2-yl, morpholin-3-yl and morpholin-4-yl. The term “thiomorpholinyl” as used herein includes thiomorpholin-2-yl, thiomorpholin-3-yl and thiomorpholin-4-yl. The term “dioxanyl” as used herein includes 1,2-dioxan-3-yl, 1,2-dioxan-4-yl, 1,3-dioxan-2-yl, 1,3-dioxan-4-yl, 1,3-dioxan-5-yl and 1,4-dioxan-2-yl. The term “dithianyl” as used herein includes 1,2-dithian-3-yl, 1,2-dithian-4-yl, 1,3-dithian-2-yl, 1,3-dithian-4-yl, 1,3-dithian-5-yl and 1,4-dithian-2-yl. The term “oxathianyl” as used herein includes oxathian-2-yl and oxathian-3-yl. The term “trioxanyl” as used herein includes 1,2,3-trioxan-4-yl, 1,2,3-trioxan-5-yl, 1,2,4-trioxan-3-yl, 1,2,4-trioxan-5-yl, 1,2,4-trioxan-6-yl and 1,3,4-trioxan-2-yl. The term “azepanyl” as used herein includes azepan-1-yl, azepan-2-yl, azepan-3-yl and azepan-4-yl. The term “homopiperazinyl” as used herein includes homopiperazin-1-yl, homopiperazin-2-yl, homopiperazin-3-yl and homopiperazin-4-yl. The term “indolinyl” as used herein includes indolin-1-yl, indolin-2-yl, indolin-3-yl, indolin-4-yl, indolin-5-yl, indolin-6-yl, and indolin-7-yl. The term “quinolizinyl” as used herein includes quinolizidin-1-yl, quinolizidin-2-yl, quinolizidin-3-yl and quinolizidin-4-yl. The term “isoindolinyl” as used herein includes isoindolin-1-yl, isoindolin-2-yl, isoindolin-3-yl, isoindolin-4-yl, isoindolin-5-yl, isoindolin-6-yl, and isoindolin-7-yl. The term “3H-indolyl” as used herein includes 3H-indol-2-yl, 3H-indol-3-yl, 3H-indol-4-yl, 3H-indol-5-yl, 3H-indol-6-yl, and 3H-indol-7-yl. The term “quinolizinyl” as used herein includes quinolizidin-1-yl, quinolizidin-2-yl, quinolizidin-3-yl and quinolizidin-4-yl. The term “quinolizinyl” as used herein includes quinolizidin-1-yl, quinolizidin-2-yl, quinolizidin-3-yl and quinolizidin-4-yl. The term “tetrahydroquinolinyl” as used herein includes tetrahydroquinolin-1-yl, tetrahydroquinolin-2-yl, tetrahydroquinolin-3-yl, tetrahydroquinolin-4-yl, tetrahydroquinolin-5-yl, tetrahydroquinolin-6-yl, tetrahydroquinolin-7-yl and tetrahydroquinolin-8-yl. The term “tetrahydroisoquinolinyl” as used herein includes tetrahydroisoquinolin-1-yl, tetrahydroisoquinolin-2-yl, tetrahydroisoquinolin-3-yl, tetrahydroisoquinolin-4-yl, tetrahydroisoquinolin-5-yl, tetrahydroisoquinolin-6-yl, tetrahydroisoquinolin-7-yl and tetrahydroisoquinolin-8-yl. The term “chromanyl” as used herein includes chroman-2-yl, chroman-3-yl, chroman-4-yl, chroman-5-yl, chroman-6-yl, chroman-7-yl and chroman-8-yl. The term “1H-pyrrolizine” as used herein includes 1H-pyrrolizin-1-yl, 1H-pyrrolizin-2-yl, 1H-pyrrolizin-3-yl, 1H-pyrrolizin-5-yl, 1H-pyrrolizin-6-yl and 1H-pyrrolizin-7-yl. The term “3H-pyrrolizine” as used herein includes 3H-pyrrolizin-1-yl, 3H-pyrrolizin-2-yl, 3H-pyrrolizin-3-yl, 3H-pyrrolizin-5-yl, 3H-pyrrolizin-6-yl and 3H-pyrrolizin-7-yl.


The term “heteroaryl” refers to an aromatic ring system of 5 to 18 atoms including at least one N, O, S, or P, containing 1 or 2 rings which can be fused together or linked covalently, each ring typically containing 5 to 6 atoms; at least one of said rings is aromatic, where the N and S heteroatoms may optionally be oxidized and the N heteroatoms may optionally be quaternized, and wherein at least one carbon atom of said heteroaryl can be oxidized to form at least one C═O. Fused systems of a heteroaryl ring with a cycloalkyl ring, or a cycloalkenyl ring, or a cycloalkynyl ring, are considered as heteroaryl irrespective of the ring that is bound to the core structure. Fused systems of a heteroaryl ring with a heterocycle are considered as heteroaryl irrespective of the ring that is bound to the core structure. Fused systems of a hetero aryl ring with an aryl ring are considered as heteroaryl irrespective of the ring that is bound to the core structure. Non-limiting examples of such heteroaryl, include: triazol-2-yl, pyridinyl, 1H-pyrazol-5-yl, pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, oxatriazolyl, thiatriazolyl, pyrimidyl, pyrazinyl, pyridazinyl, oxazinyl, dioxinyl, thiazinyl, triazinyl, imidazo[2,1-b][1,3]thiazolyl, thieno[3,2-b]furanyl, thieno[3,2-b]thiophenyl, thieno[2,3-d][1,3]thiazolyl, thieno[2,3-d]imidazolyl, tetrazolo[1,5-a]pyridinyl, indolyl, indolizinyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzothiophenyl, isobenzothiophenyl, indazolyl, benzimidazolyl, 1,3-benzoxazolyl, 1,2-benzisoxazolyl, 2,1-benzisoxazolyl, 1,3-benzothiazolyl, 1,2-benzoisothiazolyl, 2,1-benzoisothiazolyl, benzotriazolyl, 1,2,3-benzoxadiazolyl, 2,1,3-benzoxadiazolyl, 1,2,3-benzothiadiazolyl, 2,1,3-benzothiadiazolyl, benzo[d]oxazol-2(3H)-one, 2,3-dihydro-benzofuranyl, thienopyridinyl, purinyl, imidazo[1,2-a]pyridinyl, 6-oxo-pyridazin-1(6H)-yl, 2-oxopyridin-1(2H)-yl, 6-oxo-pyridazin-1(6H)-yl, 2-oxopyridin-1(2H)-yl, 1,3-benzodioxolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, quinoxalinyl; in some embodiments, said heteroaryl group is selected from the group comprising pyridyl, pyrazinyl, pyrimidinyl, pyrazolyl, pyrrolyl, isoxazolyl, thiophenyl, imidazolyl, indolyl, benzimidazolyl, s-triazinyl, oxazolyl, isothiazolyl, furyl, thienyl, triazolyl and thiazolyl; in some embodiments, said heteroaryl group is selected from the group comprising pyridyl, pyrazinyl, pyrimidinyl, indolyl and benzimidazolyl.


The term “pyrrolyl” (also called azolyl) as used herein includes pyrrol-1-yl, pyrrol-2-yl and pyrrol-3-yl. The term “furanyl” (also called “furyl”) as used herein includes furan-2-yl and furan yl (also called furan-2-yl and furan-3-yl). The term “thiophenyl” (also called “thienyl”) as used herein includes thiophen-2-yl and thiophen-3-yl (also called thien-2-yl and thien-3-yl). The term “pyrazolyl” (also called 1H-pyrazolyl and 1,2-diazolyl) as used herein includes pyrazol-1-yl, pyrazol-3-yl or 1H-pyrazol-5-yl, pyrazol-4-yl and pyrazol-5-yl. The term “imidazolyl” as used herein includes imidazol-1-yl, imidazol-2-yl, imidazol-4-yl and imidazol-5-yl. The term “oxazolyl” (also called 1,3-oxazolyl) as used herein includes oxazol-2-yl, oxazol-4-yl and oxazol-5-yl. The term “isoxazolyl” (also called 1,2-oxazolyl), as used herein includes isoxazol-3-yl, isoxazol-4-yl, and isoxazol-5-yl. The term “thiazolyl” (also called 1,3-thiazolyl), as used herein includes thiazol-2-yl, thiazol-4-yl and thiazol-5-yl (also called 2-thiazolyl, 4-thiazolyl and 5-thiazolyl). The term “isothiazolyl” (also called 1,2-thiazolyl) as used herein includes isothiazol-3-yl, isothiazol-4-yl, and isothiazol-5-yl. The term “triazolyl” as used herein includes triazol-2-yl, 1H-triazolyl and 4H-1,2,4-triazolyl, “1H-triazolyl” includes 1H-1,2,3-triazol-1-yl, 1H-1,2,3-triazol-4-yl, 1H-1,2,3-triazol-5-yl, 1H-1,2,4-triazol-1-yl, 1H-1,2,4-triazol-3-yl and 1H-1,2,4-triazol-5-yl. “4H-1,2,4-triazolyl” includes 4H-1,2,4-triazol-4-yl, and 4H-1,2,4-triazol-3-yl. The term “oxadiazolyl” as used herein includes 1,2,3-oxadiazol-4-yl, 1,2,3-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, 1,2,4-oxadiazol-5-yl, 1,2,5-oxadiazol-3-yl and 1,3,4-oxadiazol-2-yl. The term “thiadiazolyl” as used herein includes 1,2,3-thiadiazol-4-yl, 1,2,3-thiadiazol-5-yl, 1,2,4-thiadiazol-3-yl, 1,2,4-thiadiazol-5-yl, 1,2,5-thiadiazol-3-yl (also called furazan-3-yl) and 1,3,4-thiadiazol-2-yl. The term “tetrazolyl” as used herein includes 1H-tetrazol-1-yl, 1H-tetrazol-5-yl, 2H-tetrazol-2-yl, and 2H-tetrazol-5-yl. The term “oxatriazolyl” as used herein includes 1,2,3,4-oxatriazol-5-yl and 1,2,3,5-oxatriazol-4-yl. The term “thiatriazolyl” as used herein includes 1,2,3,4-thiatriazol-5-yl and 1,2,3,5-thiatriazol-4-yl. The term “pyridinyl” (also called “pyridyl”) as used herein includes pyridin-2-yl, pyridin-3-yl and pyridin-4-yl (also called 2-pyridyl, 3-pyridyl and 4-pyridyl). The term “pyrimidyl” as used herein includes pyrimid-2-yl, pyrimid-4-yl, pyrimid-5-yl and pyrimid-6-yl. The term “pyrazinyl” as used herein includes pyrazin-2-yl and pyrazin-3-yl. The term “pyridazinyl as used herein includes pyridazin-3-yl and pyridazin-4-yl. The term “oxazinyl” (also called “1,4-oxazinyl”) as used herein includes 1,4-oxazin-4-yl and 1,4-oxazin-5-yl. The term “dioxinyl” (also called “1,4-dioxinyl”) as used herein includes 1,4-dioxin-2-yl and 1,4-dioxin-3-yl. The term “thiazinyl” (also called “1,4-thiazinyl”) as used herein includes 1,4-thiazin-2-yl, 1,4-thiazin-3-yl, 1,4-thiazin-4-yl, 1,4-thiazin-5-yl and 1,4-thiazin-6-yl. The term “triazinyl” as used herein includes 1,3,5-triazin-2-yl, 1,2,4-triazin-3-yl, 1,2,4-triazin-5-yl, 1,2,4-triazin-6-yl, 1,2,3-triazin-4-yl and 1,2,3-triazin-5-yl. The term “imidazo[2,1-b][1,3]thiazolyl” as used herein includes imidazo[2,1-b][1,3]thiazoi-2-yl, imidazo[2,1-b][1,3]thiazol-3-yl, imidazo[2,1-b][1,3]thiazol-5-yl and imidazo[2,1-b][1,3]thiazol-6-yl. The term “thieno[3,2-b]furanyl” as used herein includes thieno[3,2-b]furan-2-yl, thieno[3,2-b]furan-3-yl, thieno[3,2-b]furan-4-yl, and thieno[3,2-b]furan-5-yl. The term “thieno[3,2-b]thiophenyl” as used herein includes thieno[3,2-b]thien-2-yl, thieno[3,2-b]thien-3-yl, thieno[3,2-b]thien-5-yl and thieno[3,2-b]thien-6-yl. The term “thieno[2,3-d][1,3]thiazolyl” as used herein includes thieno[2,3-d][1,3]thiazol-2-yl, thieno[2,3-d][1,3]thiazol-5-yl and thieno[2,3-d][1,3]thiazol-6-yl. The term “thieno[2,3-d]imidazolyl” as used herein includes thieno[2,3-d]imidazol-2-yl, thieno[2,3-d]imidazol-4-yl and thieno[2,3-d]imidazol-5-yl. The term “tetrazolo[1,5-a]pyridinyl” as used herein includes tetrazolo[1,5-a]pyridine-5-yl, tetrazolo[1,5-a]pyridine-6-yl, tetrazolo[1,5-a]pyridine-7-yl, and tetrazolo[1,5-a]pyridine-8-yl. The term “indolyl” as used herein includes indol-1-yl, indol-2-yl, indol-3-yl, -indol-4-yl, indol-5-yl, indol-6-yl and indol-7-yl. The term “indolizinyl” as used herein includes indolizin-1-yl, indolizin-2-yl, indolizin-3-yl, indolizin-5-yl, indolizin-6-yl, indolizin-7-yl, and indolizin-8-yl. The term “isoindolyl” as used herein includes isoindol-1-yl, isoindol-2-yl, isoindol-3-yl, isoindol-4-yl, isoindol-5-yl, isoindol-6-yl and isoindol-7-yl. The term “benzofuranyl” (also called benzo[b]furanyl) as used herein includes benzofuran-2-yl, benzofuran-3-yl, benzofuran-4-yl, benzofuran-5-yl, benzofuran-6-yl and benzofuran-7-yl. The term “isobenzofuranyl” (also called benzo[c]furanyl) as used herein includes isobenzofuran-1-yl, isobenzofuran-3-yl, isobenzofuran-4-yl, isobenzofuran-5-yl, isobenzofuran-6-yl and isobenzofuran-7-yl. The term “benzothiophenyl” (also called benzo[b]thienyl) as used herein includes 2-benzo[b]thiophenyl, 3-benzo[b]thiophenyl, 4-benzo[b]thiophenyl, 5-benzo[b]thiophenyl, 6-benzo[b]thiophenyl and -7-benzo[b]thiophenyl (also called benzothien-2-yl, benzothien-3-yl, benzothien-4-yl, benzothien-5-yl, benzothien-6-yl and benzothien-7-yl). The term “isobenzothiophenyl” (also called benzo[c]thienyl) as used herein includes isobenzothien-1-yl, isobenzothien-3-yl, isobenzothien-4-yl, isobenzothien-5-yl, isobenzothien-6-yl and isobenzothien-7-yl. The term “indazolyl” (also called 1H-indazolyl or 2-azaindolyl) as used herein includes 1H-indazol-1-yl, 1H-indazol-3-yl, 1H-indazol-4-yl, 1H-indazol-5-yl, 1H-indazol-6-yl, 1H-indazol-7-yl, 2H-indazol-2-yl, 2H-indazol-3-yl, 2H-indazol-4-yl, 2H-indazol-5-yl, 2H-indazol-6-yl, and 2H-indazol-7-yl. The term “benzimidazolyl” as used herein includes benzimidazol-1-yl, benzimidazol-2-yl, benzimidazol-4-yl, benzimidazol-5-yl, benzimidazol-6-yl and benzimidazol-7-yl. The term “1,3-benzoxazolyl” as used herein includes 1,3-benzoxazol-2-yl, 1,3-benzoxazol-4-yl, 1,3-benzoxazol-5-yl, 1,3-benzoxazol-6-yl and 1,3-benzoxazol-7-yl. The term “1,2-benzisoxazolyl” as used herein includes 1,2-benzisoxazol-3-yl, 1,2-benzisoxazol-4-yl, 1,2-benzisoxazol-5-yl, 1,2-benzisoxazol-6-yl and 1,2-benzisoxazol-7-yl. The term “2,1-benzisoxazolyl” as used herein includes 2,1-benzisoxazol-3-yl, 2,1-benzisoxazol-4-yl, 2,1-benzisoxazol-5-yl, 2,1-benzisoxazol yl and 2,1-benzisoxazol-7-yl. The term “1,3-benzothiazolyl” as used herein includes 1,3-benzothiazol-2-yl, 1,3-benzothiazol-4-yl, 1,3-benzothiazol-5-yl, 1,3-benzothiazol-6-yl and 1,3-benzothiazol-7-yl. The term “1,2-benzoisothiazolyl” as used herein includes 1,2-benzisothiazol yl, 1,2-benzisothiazol-4-yl, 1,2-benzisothiazol-5-yl, 1,2-benzisothiazol-6-yl and 1,2-benzisothiazol-7-yl. The term “2,1-benzoisothiazolyl” as used herein includes 2,1-benzisothiazol-3-yl, 2,1-benzisothiazol-4-yl, 2,1-benzisothiazol-5-yl, 2,1-benzisothiazol-6-yl and 2,1-benzisothiazol-7-yl. The term “benzotriazolyl” as used herein includes benzotriazol-1-yl, benzotriazol-4-yl, benzotriazol-5-yl, benzotriazol-6-yl and benzotriazol-7-yl. The term “1,2,3-benzoxadiazolyl” as used herein includes 1,2,3-benzoxadiazol-4-yl, 1,2,3-benzoxadiazol-5-yl, 1,2,3-benzoxadiazol-6-yl and 1,2,3-benzoxadiazol-7-yl. The term “2,1,3-benzoxadiazolyl” as used herein includes 2,1,3-benzoxadiazol-4-yl, 2,1,3-benzoxadiazol-5-yl, 2,1,3-benzoxadiazol-6-yl and 2,1,3-benzoxadiazol-7-yl. The term “1,2,3-benzothiadiazolyl” as used herein includes 1,2,3-benzothiadiazol-4-yl, 1,2,3-benzothiadiazol-5-yl, 1,2,3-benzothiadiazol-6-yl and 1,2,3-benzothiadiazol-7-yl. The term “2,1,3-benzothiadiazolyl” as used herein includes 2,1,3-benzothiadiazol-4-yl, 2,1,3-benzothiadiazol-5-yl, 2,1,3-benzothiadiazol-6-yl and 2,1,3-benzothiadiazol-7-yl. The term “thienopyridinyl” as used herein includes thieno[2,3-b]pyridinyl, thieno[2,3-c]pyridinyl, thieno[3,2-c]pyridinyl and thieno[3,2-b]pyridinyl. The term “purinyl” as used herein includes purin-2-yl, purin-6-yl, purin-7-yl and purin-8-yl. The term “imidazo[1,2-a]pyridinyl”, as used herein includes imidazo[1,2-a]pyridin-2-yl, imidazo[1,2-a]pyridin-3-yl, imidazo[1,2-a]pyridin-4-yl, imidazo[1,2-a]pyridin-5-yl, imidazo[1,2-a]pyridin-6-yl and imidazo[1,2-a]pyridin-7-yl. The term “1,3-benzodioxolyl”, as used herein includes 1,3-benzodioxol-4-yl, 1,3-benzodioxol-5-yl, 1,3-benzodioxol-6-yl, and 1,3-benzodioxol-7-yl. The term “quinolinyl” as used herein includes quinolin-2-yl, quinolin-3-yl, quinolin-4-yl, quinolin-5-yl, quinolin-6-yl, quinolin-7-yl and quinolin-8-yl. The term “isoquinolinyl” as used herein includes isoquinolin-1-yl, isoquinolin-3-yl, isoquinolin-4-yl, isoquinolin-5-yl, isoquinolin-6-yl, isoquinolin-7-yl and isoquinolin-8-yl. The term “cinnolinyl” as used herein includes cinnolin-3-yl, cinnolin-4-yl, cinnolin-5-yl, cinnolin-6-yl, cinnolin-7-yl and cinnolin-8-yl. The term “quinazolinyl” as used herein includes quinazolin-2-yl, quinazolin-4-yl, quinazolin-5-yl, quinazolin-6-yl, quinazolin-7-yl and quinazolin-8-yl. The term “quinoxalinyl” as used herein includes quinoxalin-2-yl, quinoxalin-5-yl, and quinoxalin-6-yl.


Heteroaryl and heterocycle or heterocyclyl as used herein includes by way of example and not limitation these groups described in Paquette, Leo A. “Principles of Modern Heterocyclic Chemistry” (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; “The Chemistry of Heterocyclic Compounds, A series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; Katritzky, Alan R., Rees, C. W. and Scriven, E. “Comprehensive Heterocyclic Chemistry” (Pergamon Press, 1996); and J. Am. Chem. Soc. (1960) 82:5566.


The term “heterocyclyloxy” or “heterocycleoxy”, as a group or part of a group, refers to a group having the formula —O—Ri wherein Ri is heterocyclyl as defined herein above.


The term “heterocyclylalkyloxy” or “heterocycleoxy”, as a group or part of a group, refers to a group having the formula —O—Ra—Ri wherein Ri is heterocyclyl, and Ra is alkyl as defined herein above.


The term “heteroaryloxy”, as a group or part of a group, refers to a group having the formula —O—Rk wherein Rk is heteroaryl as defined herein above.


The term “heteroarylalkyloxy”, as a group or part of a group, refers to a group having the formula —O—Ra—Ri wherein Ri is heteroaryl, and Ra is alkyl as defined herein above.


The term “heterocyclyl-alkyl” or “heterocycle-alkyl” as a group or part of a group, refers to an alkyl in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heterocyclyl. A non-limiting example of a heterocyclyl-alkyl or heterocycle-alkyl group is 2-piperidinyl-methylene. The heterocyclyl-alkyl or heterocycle-alkyl group can comprise 6 to 20 atoms, e.g. the alkyl moiety is 1 to 6 carbon atoms and the heterocyclyl moiety is 3 to 14 atoms.


The term “heterocyclyl-alkenyl” or “heterocycle-alkenyl” as a group or part of a group refers to an alkenyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an heterocyclyl. The heterocyclyl-alkenyl or heterocycle-alkenyl group can comprise 6 to 20 atoms, e.g. the alkenyl moiety is 2 to 6 carbon atoms and the heterocyclyl moiety is 3 to 14 atoms.


The term “heterocyclyl-alkynyl” or “heterocycle-alkynyl” as a group or part of a group refers to an alkynyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with a heterocyclyl. The heterocyclyl-alkynyl or heterocycle-alkynyl group can comprise 6 to 20 atoms, e.g. the alkynyl moiety can comprise 2 to 6 carbon atoms and the heterocyclyl moiety can comprise 3 to 14 atoms.


The term “heterocyclyl-heteroalkyl” or “heterocycle-heteroalkyl” as a group or part of a group refers to a heteroalkyl in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heterocyclyl. The heterocyclyl-heteroalkyl or heterocycle-heteroalkyl group can comprise 6 to 20 atoms, e.g. the heteroalkyl moiety can comprise 1 to 6 carbon atoms and the heterocyclyl moiety can comprise 3 to 14 atoms. In some embodiments heterocyclyl-heteroalkyl or heterocycle-heteroalkyl is selected from the group comprising heterocyclyl-O-alkyl, heterocyclylalkyl-O-alkyl, heterocyclyl-NH-alkyl, heterocyclyl-N(alkyl)2, heterocyclylalkyl-NH-alkyl, heterocyclylalkyl-N-(alkyl)2, heterocyclyl-S-alkyl, and heterocyclylalkyl-S-alkyl.


The term “heterocyclyl-heteroalkenyl” or “heterocycle-heteroalkenyl” as a group or part of a group refers to a heteroalkenyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with a heterocyclyl. The heterocyclyl-heteroalkenyl or heterocycle-heteroalkenyl group can comprise 6 to 20 atoms, e.g. the heteroalkenyl moiety can comprise 2 to 6 carbon atoms and the heterocyclyl moiety can comprise 3 to 14 atoms. In some embodiments heterocyclyl-heteroalkenyl or heterocycle-heteroalkenyl is selected from the group comprising heterocyclyl-O-alkenyl, heterocyclylalkyl-O-alkenyl, heterocyclyl-NH-alkenyl, heterocyclyl-N(alkenyl)2, heterocyclylalkyl-NH-alkenyl, heterocyclylalkyl-N-(alkenyl)2, heterocyclyl-S-alkenyl, and heterocyclylalkenyl-S-alkenyl.


The term “heterocyclyl-heteroalkynyl” or “heterocycle-heteroalkynyl” as a group or part of a group refers to a heteroalkynyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with a heterocyclyl. The heterocyclyl-heteroalkynyl or heterocycle-heteroalkynyl group can comprise 6 to 20 atoms, e.g. the heteroalkynyl moiety can comprise 2 to 6 carbon atoms and the heterocyclyl moiety can comprise 3 to 14 atoms. In some embodiments heterocyclyl-heteroalkynyl is selected from the group comprising heterocyclyl-O-alkynyl, heterocyclylalkynyl-O-alkynyl, heterocyclyl-NH-alkynyl, heterocyclyl-N(alkynyl)2, heterocyclylalkynyl-NH-alkynyl, heterocyclylalkynyl-N-(alkynyl)2, heterocyclyl-S-alkynyl, and heterocyclylalkynyl-S-alkynyl.


The term “heteroaryl-alkyl” as a group or part of a group refers to an alkyl in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heteroaryl. An example of a heteroaryl-alkyl group is 2-pyridyl-methylene. The heteroaryl-alkyl group can comprise 6 to 20 atoms, e.g. the alkyl moiety of the heteroaryl-alkyl group can comprise 1 to 6 carbon atoms and the heteroaryl moiety can comprise 5 to 14 atoms.


The term “heteroaryl-alkenyl” as a group or part of a group refers to an alkenyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an heteroaryl. The heteroaryl-alkenyl group can comprise 6 to 20 atoms, e.g. the alkenyl moiety of the heteroaryl-alkenyl group can comprise 2 to 6 carbon atoms and the heteroaryl moiety can comprise 5 to 14 atoms.


The term “heteroaryl-alkynyl” as a group or part of a group as used herein refers to an alkynyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with a heteroaryl. The heteroaryl-alkynyl group comprises 6 to 20 atoms, e.g. the alkynyl moiety of the heteroaryl-alkynyl group is 2 to 6 carbon atoms and the heteroaryl moiety is 5 to 14 atoms.


The term “heteroaryl-heteroalkyl” as a group or part of a group as used herein refers to a heteroalkyl in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heteroaryl. The heteroaryl-heteroalkyl group comprises 7 to 20 atoms, e.g. the heteroalkyl moiety of the heteroaryl-heteroalkyl group is 2 to 6 carbon atoms and the heteroaryl moiety is 5 to 14 atoms. In some embodiments heteroaryl-heteroalkyl is selected from the group comprising heteroaryl-O-alkyl, heteroarylalkyl-O-alkyl, heteroaryl-NH-alkyl, heteroaryl-N(alkyl)2, heteroarylalkyl-NH-alkyl, heteroarylalkyl-N-(alkyl)2, heteroaryl-S-alkyl, and heteroarylalkyl-S-alkyl.


The term “heteroaryl-heteroalkenyl” as a group or part of a group as used herein refers to a heteroalkenyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with an heteroaryl. The heteroaryl-heteroalkenyl group comprises 8 to 20 atoms, e.g. the heteroalkenyl moiety of the heteroaryl-heteroalkenyl group is 3 to 6 carbon atoms and the heteroaryl moiety is 5 to 14 atoms. In some embodiments heteroaryl-heteroalkenyl is selected from the group comprising heteroaryl-O-alkenyl, heteroarylalkenyl-O-alkenyl, heteroaryl-NH-alkenyl, heteroaryl-N(alkenyl)2, heteroarylalkenyl-NH-alkenyl, heteroarylalkenyl-N-(alkenyl)2, heteroaryl-S-alkenyl, and heteroarylalkenyl-S-alkenyl.


The term “heteroaryl-heteroalkynyl” as a group or part of a group as used herein refers to a heteroalkynyl in which one of the hydrogen atoms bonded to a carbon atom, is replaced with a heteroaryl. The heteroaryl-heteroalkynyl group comprises 8 to 20 atoms, e.g. the heteroalkynyl moiety of the heteroaryl-heteroalkynyl group is 2 to 6 carbon atoms and the heteroaryl moiety is 5 to 14 atoms. In some embodiments heteroaryl-heteroalkynyl is selected from the group comprising heteroaryl-O-alkynyl, heteroarylalkynyl-O-alkynyl, heteroaryl-NH-alkynyl, heteroaryl-N(alkynyl)2, heteroarylalkynyl-NH-alkynyl, heteroarylalkynyl-N-(alkynyl)2, heteroaryl-S-alkynyl, and heteroarylalkynyl-S-alkynyl.


By way of example, carbon bonded heteroaryl or heterocyclic rings (or heterocycles) can be bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heteroaryls and heterocyclyls include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl. By way of example, nitrogen bonded heterocyclic rings are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ß-carboline. Still more typically, nitrogen bonded heteroaryls or heterocyclyls include 1-aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1-pyrazolyl, and 1-piperidinyl.


As used herein and unless otherwise stated, the terms “alkoxy”, “cyclo-alkoxy”, “aryloxy”, “arylalkyloxy”, “heteroaryloxy” “heterocyclyloxy”, “alkylthio”, “cycloalkylthio”, “arylthio”, “arylalkylthio”, “heteroarylthio” and “heterocyclylthio” refer to substituents wherein an alkyl group, respectively a cycloalkyl, aryl, arylalkyl heteroaryl, or heterocyclyl (each of them such as defined herein), are attached to an oxygen atom or a sulfur atom through a single bond, such as but not limited to methoxy, ethoxy, propoxy, butoxy, thioethyl, thiomethyl, phenyloxy, benzyloxy, mercaptobenzyl and the like. The same definitions will apply for alkenyl and alkynyl instead of alkyl.


The term “alkylthio”, as a group or part of a group, refers to a group having the formula —S—Rb wherein Rb is alkyl as defined herein above. Non-limiting examples of alkylthio groups include methylthio (—SCH3), ethylthio (—SCH2CH3), n-propylthio, isopropylthio, n-butylthio, isobutylthio, sec-butylthio, tert-butylthio and the like.


The term “alkenylthio”, as a group or part of a group, refers to a group having the formula —S—Rd wherein Rd is alkenyl as defined herein above.


The term “alkynylthio”, as a group or part of a group, refers to a group having the formula —S—Re wherein Re is alkynyl as defined herein above.


The term “arylthio”, as a group or part of a group, refers to a group having the formula —S—Rg wherein Rg is aryl as defined herein above.


The term “arylalkylthio”, as a group or part of a group, refers to a group having the formula —S—Ra—Rg wherein Ra is alkylene and Rg is aryl as defined herein above.


The term “heterocyclylthio”, as a group or part of a group, refers to a group having the formula —S—Ri wherein Ri is heterocyclyl as defined herein above.


The term “heteroarylthio”, as a group or part of a group, refers to a group having the formula —S—Rk wherein Rk is heteroaryl as defined herein above.


The term “heterocyclylalkylthio”, as a group or part of a group, refers to a group having the formula —S—Ra—Ri wherein Ra is alkylene and Ri is heterocyclyl as defined herein above.


The term “heteroarylalkylthio”, as a group or part of a group, refers to a group having the formula —S—Ra—Rk wherein Ra is alkylene and Rk is heteroaryl as defined herein above.


The term “mono- or di-alkylamino”, as a group or part of a group, refers to a group of formula) —N(Ro(Rb) wherein Ro is hydrogen, or alkyl, Rb is alkyl. Thus, alkylamino include mono-alkyl amino group (e.g. mono-alkylamino group such as methylamino and ethylamino), and di-alkylamino group (e.g. di-alkylamino group such as dimethylamino and diethylamino). Non-limiting examples of suitable mono- or di-alkylamino groups include n-propylamino, isopropylamino, n-butylamino, i-butylamino, sec-butylamino, t-butylamino, pentylamino, n-hexylamino, di-n-propylamino, di-i-propylamino, ethylmethylamino, methyl-n-propylamino, methyl-i-propylamino, n-butylmethylamino, i-butylmethylamino, t-butylmethylamino, ethyl-n-propylamino, ethyl-i-propylamino, n-butylethylamino, i-butylethylamino, t-butylethylamino, di-n-butylamino, di-i-butylamino, methylpentylamino, methylhexylamino, ethylpentylamino, ethylhexylamino, propylpentylamino, propylhexylamino, and the like.


The term “mono- or di-arylamino”, as a group or part of a group, refers to a group of formula —N(Rq)(Rr) wherein Rq and Rr are each independently selected from hydrogen, aryl, or alkyl, wherein at least one of Rq or Rr is aryl.


The term “mono- or di-heteroarylamino”, as a group or part of a group, refers to a group of formula —N(Ru)(Rv) wherein Ru and Rv are each independently selected from hydrogen, heteroaryl, or alkyl, wherein at least one of Ru or Rv is heteroaryl as defined herein.


The term “mono- or di-heterocyclylamino”, as a group or part of a group, refers to a group of formula —N(Rw)(Rx) wherein Rw and Rx are each independently selected from hydrogen, heterocyclyl, or alkyl, wherein at least one of Rw or Rx is heterocyclyl as defined herein.


As used herein and unless otherwise stated, the term halogen means any atom selected from the group consisting of fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).


The terminology regarding a chemical group “which optionally includes one or more heteroatoms, said heteroatoms being selected from the atoms consisting of O, S, and N” as used herein, refers to a group where one or more carbon atoms are replaced by an oxygen, nitrogen or sulphur atom and thus includes, depending on the group to which is referred, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloheteroalkyl, cycloheteroalkenyl, cycloheteroalkynyl, heteroaryl, aryl heteroalkyl, heteroarylalkyl, heteroarylheteroalkyl, arylheteroalkenyl, heteroarylalkenyl, heteroarylheteroalkenyl, heteroarylheteroalkenyl, arylheteroalkynyl, heteroarylalkynyl, heteroarylheteroalkynyl, among others. This term therefore comprises, depending on the group to which is referred, as an example alkoxy, alkenyloxy, alkynyloxy, alkyl-O-alkylene, alkenyl-O-alkylene, arylalkoxy, benzyloxy, heteroaryl-heteroalkyl, heterocyclyl-heteroalkyl, heteroaryl-alkoxy, heterocyclyl-alkoxy, among others. As an example, the terminology “alkyl which optionally includes one or more heteroatoms, said heteroatoms being selected from the atoms consisting of O, S, and N” therefore refers to heteroalkyl, meaning an alkyl which comprises one or more heteroatoms in the hydrocarbon chain, whereas the heteroatoms may be positioned at the beginning of the hydrocarbon chain, in the hydrocarbon chain or at the end of the hydrocarbon chain. Examples of heteroalkyl include methoxy, methylthio, ethoxy, propoxy, CH3—O—CH2—, CH3—S—CH2—, CH3—CH2—O—CH2—, CH3—NH—, (CH3)2—N—, (CH3)2—CH2—NH—CH2—CH2—, among many other examples. As an example, the terminology “arylalkylene which optionally includes one or more heteroatoms in the alkylene chain, said heteroatoms being selected from the atoms consisting of O, S, and N” therefore refers to arylheteroalkylene, meaning an arylalkylene which comprises one or more heteroatoms in the hydrocarbon chain, whereas the heteroatoms may be positioned at the beginning of the hydrocarbon chain, in the hydrocarbon chain or at the end of the hydrocarbon chain. “Arylheteroalkylene” thus includes aryloxy, arylalkoxy, aryl-alkyl-NH— and the like and examples are phenyloxy, benzyloxy, aryl-CH2—S—CH2—, aryl-CH2—O—CH2—, aryl-NH—CH2— among many other examples. The same counts for “heteroalkenylene”, “heteroalkynylene”, and other terms used herein when referred to “which optionally includes one or more heteroatoms, said heteroatoms being selected from the atoms consisting of O, S, and N”.


The terminology regarding a chemical group “wherein optionally two or more hydrogen atoms on a carbon atom or heteroatom of said group can be taken together to form a ═O or ═S” as used herein, refers to a group where two or more hydrogen atoms on a carbon atom or heteroatom of said group are taken together to form ═O or ═S. As an example, the terminology refers to “an alkyl wherein optionally two or more hydrogen atoms on a carbon atom or heteroatom of said alkyl can be taken together to form a ═O or ═S”, includes among other examples CH3—C(O)—CH2—, CH3—C(O)—, CH3—C(S)—CH2—, CH3—S(O)2—CH2— and (CH3)2—CH2—C(O)—CH2—CH2—.


The combination for a group “which optionally includes one or more heteroatoms, said heteroatoms being selected from the atoms consisting of O, S, and N” and “wherein optionally two or more hydrogen atoms on a carbon atom or heteroatom of said group can be taken together to form a ═O or ═S” can combine the two aspects described herein above and includes, if the group referred to is alkyl, among other examples CH3—C(O)O—, CH3—C(O)O—CH2—, CH3—NH—C(O)—, CH3—C(O)—NH—CH3—NH—C(O)—CH2—, CH3—NH—C(S)—CH2—, CH3—NH—C(S)—NH—CH2—, CH3—NH—S(O)2— and CH3—NH—S(O)2—NH—CH2—.


The term “single bond” as used herein for a linking group i.e. in a way that a certain linking group is selected from a single bond, etc. in the formulas herein, refers to a molecule wherein the linking group is not present and therefore refers to compounds with a direct linkage via a single bond between the two moieties being linked by the linking group.


As used herein with respect to a substituting group, and unless otherwise stated, the terms “substituted” such as in “substituted alkyl”, “substituted alkenyl”, substituted alkynyl”, “substituted aryl”, “substituted heteroaryl”, “substituted heterocyclyl”, “substituted arylalkyl”, “substituted heteroaryl-alkyl”, “substituted heterocyclyl-alkyl” and the like refer to the chemical structures defined herein, and wherein the said alkyl, alkenyl, alkynyl, group and/or the said aryl, heteroaryl, or heterocyclyl may be optionally substituted with one or more substituents (preferable 1, 2, 3, 4, 5 or 6), meaning that one or more hydrogen atoms are each independently replaced with at least one substituent. Typical substituents include, but are not limited to and in a particular embodiment said substituents are being independently selected from the group consisting of halogen, amino, hydroxyl, sulfhydryl, alkyl, alkoxy, alkenyl, alkenyloxy, alkynyl, alkynyloxy, cycloalkyl, cycloalkenyl, cycloalkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, heterocyclyl, arylalkyl, arylalkenyl, arylalkynyl, cycloalkyl-alkyl, cycloalkylalkenyl, cycloalkylalkynyl, heteroaryl-alkyl, heterocyclyl-alkyl, heteroaryl-alkenyl, heterocyclyl-alkenyl and heteroaryl-alkynyl, heterocyclyl-alkynyl, —X, —Z, —O, —OZ, ═O, —SZ, —S, ═S, —NZ2, —N+Z3, ═NZ, ═N—OZ, —CX3 (e.g. trifluoromethyl), —CN, —OCN, —SCN, —N═C═O, —N═C═S, —NO, —NO2, ═N2, —N3, —NZC(O)Z, —NZC(S)Z, —NZC(O)O, —NZC(O)OZ, —NZC(S)OZ, —NZC(O)NZZ, NZC(NZ)Z, NZC(NZ)NZZ, —C(O)NZZ, —C(NZ)Z, —S(O)2O, —S(O)2OZ, —S(O)2Z, —OS(O)2OZ, —OS(O)2Z, —OS(O)2O, —S(O)2NZZ, —S(O)(NZ)Z, —S(O)Z, —OP(O)(OZ)2, —P(O)(OZ)2, —P(O)(O)2, —P(O)(OZ)(O), —P(O)(OH)2, —C(O)Z, —C(O)X, —C(S)Z, —C(O)OZ, —C(O)O, —C(S)OZ, —C(O)SZ, —C(S)SZ, —C(O)NZZ, —C(S)NZZ, —C(NZ)NZZ, —OC(O)Z, —OC(S)Z, —OC(O)O, —OC(O)OZ, —OC(S)OZ, wherein each X is independently a halogen selected from F, Cl, Br, or I; and each Z is independently —H, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, protecting group or prodrug moiety, while two Z bonded to a nitrogen atom can be taken together with the nitrogen atom to which they are bonded to form a heteroaryl, or heterocyclyl. Alkyl(ene), alkenyl(ene), and alkynyl(ene) groups may also be similarly substituted.


Any substituent designation that is found in more than one site in a compound of this disclosure shall be independently selected.


Substituents optionally are designated with or without bonds. Regardless of bond indications, if a substituent is polyvalent (based on its position in the structure referred to), then any and all possible orientations of the substituent are intended.


As used herein and unless otherwise stated, the term “solvate” includes any combination which may be formed by a derivative of this disclosure with a suitable inorganic solvent (e.g. hydrates) or organic solvent, such as but not limited to alcohols, ketones, esters, ethers, nitriles and the like.


The term “heteroatom(s)” as used herein means an atom selected from nitrogen, which can be quaternized; oxygen; and sulfur, including sulfoxide and sulfone.


The term “hydroxy” as used herein means —OH.


The term “carbonyl” as used herein means carbon atom bonded to oxygen with a double bond, i.e., C═O.


The term “amino” as used herein means the —NH2 group.


The present disclosure provides novel compounds which have been shown to possess YAP/TAZ-TEAD transcription inhibitory activity. The present disclosure furthermore demonstrates that these compounds efficiently inhibit TEAD activation and thereby inhibit YAP/TAZ-TEAD transcription activation. Therefore, these compounds constitute a useful class of new potent compounds that can be used in the treatment and/or prevention of YAP/TAZ-TEAD activation mediated diseases in subjects, more specifically for the treatment and/or prevention of cancer and fibrosis, among other diseases.


The present disclosure furthermore relates to the compounds for use as medicines and to their use for the manufacture of medicaments for treating and/or preventing cancer or fibrosis.


The present disclosure relates to the compounds for use as medicines for treating and/or preventing YAP/TAZ-TEAD activation mediated diseases such as cancer or fibrosis in animals, mammals, more in particular in humans. The disclosure also relates to methods for the preparation of all such compounds and to pharmaceutical compositions comprising them in an effective amount. The present disclosure also relates to a method of treatment or prevention of cancer or fibrosis in humans by the administration of one or more such compounds, optionally in combination with one or more other medicines, to a patient in need thereof. The present disclosure also relates to the compounds for veterinary use and to their use as medicines for the prevention or treatment of diseases in a non-human mammal, such as cancer and fibrosis in non-human mammals.


In one embodiment, the disclosure provides a compound of Formula I:




embedded image


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


R1 is selected from the group consisting of:

    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl,
        • (6) —OZ1,
        • (7) C2-C6 alkenyl, and
        • (8) C2-C6 alkynyl,
      • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1,
      • (c) C2-C6 alkynyl, and
      • (d) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, C2-C8 alkenyl, and C1-C6 alkyl,
    • (iii) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (iv) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (v) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vi) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vii) —S(═O)2Z2, and
    • (viii) —C(═O)Z2;


Y1 is selected from the group consisting of ═CR2—, ═N—, and —C(═O)—;


with the provisos that:

    • (i) custom-character represents a double bond when Y1 is ═CR2— or ═N—, and R4 is absent; or
    • (ii) custom-character represents a single bond when Y1 is —C(═O)—, and R4 is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R2 is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)n—NR5aR5b;


      n is 0 or 1;


      R5a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2, and
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4, (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R5b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R3 and R4 are independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)p—NR6aR6b;


      p is 0 or 1;


      R6a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2,
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4, and
      • (x) C2-C6 alkenyl,
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R6b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      each Z1 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, C3-C8 cycloalkyl, —C(═O)OH —S(═O)2Z8, —NZ3Z4, 4- to 8-membered heterocycle, 5- to 9-membered heteroaryl, and unsubstituted or substituted aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 haloalkyl, and cyano,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl,
    • (vii) C1-C6 haloalkyl,
    • (viii) unsubstituted or substituted 4- to 8-membered heterocycle, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C2-C6 alkenyl, and
    • (ix) unsubstituted or substituted aryl, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle;


      each Z2 is independently selected from the group consisting of:
    • (i) C1-C6 alkyl,
    • (ii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) C2-C6 alkynyl,
    • (iv) C3-C6 cycloalkyl,
    • (v) C3-C6 cycloalkenyl, and
    • (vi) C1-C6 haloalkyl;


      each Z3 is independently selected from the group consisting of:
    • (i) hydrogen;
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of C2-C6 alkenyl, cyano, —C(═O)OH, —S(═O)2Z8, halogen, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z4, Z5, Z6 and Z7 is independently selected from the group consisting of:


(i) hydrogen,


(ii) C1-C6 alkyl, and


(iii) C3-C6 cycloalkyl;


each Z8 is independently selected from the group consisting of:

    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, and C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) halogen, and
    • (v) hydroxy;
    • X is selected from the group consisting of ═N— and ═CR7a—;
    • X1 is selected from the group consisting of ═N— and ═CR7b—;
    • X2 is selected from the group consisting of ═N— and ═CR7c—;
    • X3 is selected from the group consisting of ═N— and ═CR7d—; and
    • R7a, R7b, R7c, and R7d is independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, —OZ1, and —(CH2)q—NR6aR6b;


q is 0 or 1.


In another embodiment, the disclosure provides a compound of Formula II:




embedded image


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


R1 is selected from the group consisting of:

    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl,
        • (6) —OZ1,
        • (7) C2-C6 alkenyl, and
        • (8) C2-C6 alkynyl,
      • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1,
      • (c) C2-C6 alkynyl,
    • (iii) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (iv) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (v) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vi) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vii) —S(═O)2Z2, and
    • (viii) —C(═O)Z2;


Y1 is selected from the group consisting of ═CR2—, ═N—, and —C(═O)—;


with the provisos that:

    • (i) custom-character represents a double bond when Y1 is ═CR2— or ═N—, and R4 is absent; or


(ii) custom-character represents a single bond when Y1 is —C(═O)—, and R4 is selected from the group consisting of hydrogen and C1-C6 alkyl;


R2 is selected from the group consisting of:

    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)n—NR5aR5b;


      n is 0 or 1;


      R5a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) —C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2, and
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4,
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R5b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R3 and R4 are independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)p—NR6aR6b;


      p is 0 or 1;


      R6a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2, and
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4,
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R6b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      each Z1 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z2 is independently selected from the group consisting of:
    • (i) C1-C6 alkyl,
    • (ii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) C2-C6 alkynyl,
    • (iv) C3-C6 cycloalkyl,
    • (v) C3-C6 cycloalkenyl, and
    • (vi) C1-C6 haloalkyl;


      each Z3 is independently selected from the group consisting of:
    • (i) hydrogen;
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z4, Z5, Z6 and Z7 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) C3-C6 cycloalkyl;


      each Z8 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) halogen, and
    • (v) hydroxy;
    • X is selected from the group consisting of ═N— and ═CR7a—;
    • X1 is selected from the group consisting of ═N— and ═CR7b—;
    • X2 is selected from the group consisting of ═N— and ═CR7c—;
    • X3 is selected from the group consisting of ═N— and ═CR7d—; and
    • R7a, R7b, R7c, and R7d is independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, —OZ1, and —NZ3Z4.


In another embodiment, the disclosure provides a compound of Formula III:




embedded image


or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R1, R2, R3, X, X1, X2, and X3 are as defined in connection with Formula I or II.


In another embodiment, the disclosure provides a compound of Formula III, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R2 is —(CH2)n—NR5aR5b; and


R3 is selected from the group consisting of hydrogen and C1-C6 alkyl.


In another embodiment, the disclosure provides a compound of Formula III, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R5a is —C(═O)Z2 and R5b is hydrogen.


In another embodiment, the disclosure provides a compound of Formula III, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R2 is selected from the group consisting of hydrogen and C1-C6 alkyl; and


R3 is —(CH2)p—NR6aR6b.


In another embodiment, the disclosure provides a compound of Formula III, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R6a is —C(═O)Z2 and R6b is hydrogen.


In another embodiment, the disclosure provides a compound of Formula IV:




embedded image


or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R1, R3, X, X2, and X3 are as defined in connection with Formula I or II.


In another embodiment, the disclosure provides a compound of Formula IV, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R3 is —(CH2)p—NR6aR6b.


In another embodiment, the disclosure provides a compound of Formula IV, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R6a is selected from the group consisting of —C(═O)Z2 and —S(═O)2Z8; and


R6b is hydrogen.


In another embodiment, the disclosure provides a compound of Formula V:




embedded image


or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R1, R3, X, X2, and X3 are as defined in connection with Formula I or II.


In another embodiment, the disclosure provides a compound of Formula V, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R3 is —(CH2)p—NR6aR6b.


In another embodiment, the disclosure provides a compound of Formula V, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R6a is selected from the group consisting of —C(═O)Z2; and


R6b is hydrogen.


In another embodiment, the disclosure provides a compound of any one of Formulae I-V, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R1 is unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, and —OZ1.


In another embodiment, the disclosure provides a compound of any one of Formulae I-V, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


X is —CR7a═;


X1 is —CR7b═;


X2 is —CR7c═; and


X3 is —CR7d═.


In another embodiment, the disclosure provides a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of:

  • N-[[5-(cyanomethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-[[rac-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol yl]methyl]prop-2-enamide;
  • N-[[5-[[(3S)-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol yl]methyl]prop-2-enamide;
  • N-[[5-[[(3R)-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol yl]methyl]prop-2-enamide;
  • N-[[5-phenoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[6-(cyclopropoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(3-methyl-2,6-dioxo-pyrimidin-4-yl)oxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-benzyloxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[1-[4-(trifluoromethyl)phenyl]pyrazolo[3,4-c]pyridin-3-yl]methyl]prop-2-enamide;
  • N-[[5-prop-2-ynoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(cyclopropylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(cyclopropylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]methanesulfonamide;
  • 2-[[[1-[4-(trifluoromethyl)phenyl]indazol-3-yl]amino]methyl]prop-2-enoic acid;
  • N-[[5-(trifluoromethyl)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[6-(trifluoromethyl)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide; and
  • N-[[1-(4,4-difluorocyclohexyl)indazol-3-yl]methyl]prop-2-enamide.


In another embodiment, the disclosure provides a compound of Formula II, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of:

  • N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acrylamide;
  • N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acetamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)propionamide;
  • N-(1-phenyl-1H-indol-3-yl)acrylamide;
  • N-(1-(4-isopropylphenyl)-1H-indol-3-yl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)acrylamide;
  • N-(1-(4-chlorophenyl)-1H-indol-3-yl)acrylamide;
  • N-(1-(3-chlorophenyl)-1H-indol-3-yl)acrylamide;
  • N-(1-([1,1′-biphenyl]-4-yl)-1H-indol-3-yl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)propionamide;
  • N-((1-(3-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide;
  • N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acrylamide;
  • N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acetamide; and
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)propionamide.


In another embodiment, the disclosure provides a compound of Formula VI:




embedded image


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


R8 is selected from the group consisting of:


(i) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:

    • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (1) halogen,
      • (2) cyano,
      • (3) C1-C6 alkyl,
      • (4) C3-C6 cycloalkyl,
      • (5) C1-C6 haloalkyl,
      • (6) —OZ1,
      • (7) C2-C6 alkenyl, and
      • (8) C2-C6 alkynyl,
    • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (1) halogen,
      • (2) cyano,
      • (3) C1-C6 alkyl,
      • (4) C3-C6 cycloalkyl,
      • (5) C1-C6 haloalkyl, and
      • (6) —OZ1,
    • (c) C2-C6 alkynyl,


(ii) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:

    • (a) halogen,
    • (b) cyano,
    • (c) C1-C6 alkyl,
    • (d) C3-C6 cycloalkyl,
    • (e) C1-C6 haloalkyl, and
    • (f) —OZ1,


(iii) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:

    • (a) halogen,
    • (b) cyano,
    • (c) C1-C6 alkyl,
    • (d) C3-C6 cycloalkyl,
    • (e) C1-C6 haloalkyl, and
    • (f) —OZ1,


(iv) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:

    • (a) halogen,
    • (b) cyano,
    • (c) C1-C6 alkyl,
    • (d) C3-C6 cycloalkyl,
    • (e) C1-C6 haloalkyl, and
    • (f) —OZ1,


(v) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of:

    • (a) halogen,
    • (b) cyano,
    • (c) C1-C6 alkyl,
    • (d) C3-C6 cycloalkyl,
    • (e) C1-C6 haloalkyl, and
    • (f) —OZ1,


(vi) —S(═O)2Z2, and


(vii) —C(═O)Z2;


X is —N(R11)—; or X is absent;


R11 is selected from the group consisting of hydrogen and C1-C6 alkyl;


R9 is selected from the group consisting of:




embedded image


R12 is selected from the group consisting of:


(i) hydrogen,


(ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:

    • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (1) halogen,
      • (2) cyano,
      • (3) C1-C6 alkyl,
      • (4) C3-C6 cycloalkyl,
      • (5) C1-C6 haloalkyl,
      • (6) —OZ1,
      • (7) C2-C6 alkenyl, and
      • (8) C2-C6 alkynyl,
    • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (1) halogen,
      • (2) cyano,
      • (3) C1-C6 alkyl,
      • (4) C3-C6 cycloalkyl,
      • (5) C1-C6 haloalkyl, and
      • (6) —OZ1, and
    • (c) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
      • (1) halogen,
      • (2) cyano,
      • (3) C1-C6 alkyl,
      • (4) C3-C6 cycloalkyl,
      • (5) C1-C6 haloalkyl, and
      • (6) —OZ1, and


(iii) C1-C6 haloalkyl;


R10 is —(CH2)q—C(═O)R13


q is 0, 1, or 2;


R13 is selected from the group consisting of —OH, OZ1, and —NZ3Z4;


each Z1 is independently selected from the group consisting of:

    • (i) hydrogen,
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z2 is independently selected from the group consisting of:
    • (i) C1-C6 alkyl,
    • (ii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) C2-C6 alkynyl,
    • (iv) C3-C6 cycloalkyl,
    • (v) C3-C6 cycloalkenyl, and
    • (vi) C1-C6 haloalkyl;


      each Z3 is independently selected from the group consisting of:
    • (i) hydrogen;
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl; and


      each Z4 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) C3-C6 cycloalkyl.


In another embodiment, the disclosure provides a compound of Formula VI, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of:

  • 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetic acid;
  • N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetamide;
  • 2-(6-phenyl-8-(1-(pyridin-3-ylmethyl)-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-2-yl)acetic acid;
  • 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetic acid; and
  • N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetamide.


Preferred or particular statements (features) and embodiments of the compounds of this disclosure are set herein below. Each statement, aspect and embodiment of the disclosure so defined may be combined with any other statement, aspect and/or embodiment, unless clearly indicated to the contrary. In particular, any feature indicated as being preferred, particular or advantageous may be combined with any other features or statements indicated as being preferred, particular or advantageous. Hereto, the present disclosure is in particular captured by any one or any combination of one or more of the below numbered statements and embodiments, with any other statement, aspect and/or embodiment (which are not numbered).


Embodiment 1. A compound of Formula I:




embedded image


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


R1 is selected from the group consisting of:

    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl,
        • (6) —OZ1,
        • (7) C2-C6 alkenyl, and
        • (8) C2-C6 alkynyl,
      • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1,
      • (c) C2-C6 alkynyl, and
      • (d) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, C2-C8 alkenyl, and C1-C6 alkyl,
    • (iii) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (iv) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (v) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vi) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vii) —S(═O)2Z2, and
    • (viii) —C(═O)Z2;


Y1 is selected from the group consisting of ═CR2—, ═N—, and —C(═O)—;


with the provisos that:

    • (i) custom-character represents a double bond when Y1 is ═CR2— or ═N—, and R4 is absent; or
    • (ii) custom-character represents a single bond when Y1 is —C(═O)—, and R4 is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R2 is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)n—NR5aR5b;


      n is 0 or 1;


      R5a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2, and
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4,
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R5b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R3 and R4 are independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)p—NR6aR6b;


      p is 0 or 1;


      R6a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2,
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4, and
      • (x) C2-C6 alkenyl
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R6b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      each Z1 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, C3-C8 cycloalkyl, —C(═O)OH —S(═O)2Z8, —NZ3Z4, 4- to 8-membered heterocycle, 5- to 9-membered heteroaryl, and unsubstituted or substituted aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 haloalkyl, and cyano,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl,
    • (vii) C1-C6 haloalkyl,
    • (viii) unsubstituted or substituted 4- to 8-membered heterocycle, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C2-C6 alkenyl, and
    • (ix) unsubstituted or substituted aryl, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle;


      each Z2 is independently selected from the group consisting of:
    • (i) C1-C6 alkyl,
    • (ii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) C2-C6 alkynyl,
    • (iv) C3-C6 cycloalkyl,
    • (v) C3-C6 cycloalkenyl, and
    • (vi) C1-C6 haloalkyl;


      each Z3 is independently selected from the group consisting of:
    • (i) hydrogen;
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of C2-C6 alkenyl, cyano, —C(═O)OH, —S(═O)2Z8, halogen, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z4, Z5, Z6 and Z7 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) C3-C6 cycloalkyl;


      each Z8 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of halogen, cyano, and C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) halogen, and
    • (v) hydroxy;
    • X is selected from the group consisting of ═N— and ═CR7a—;
    • X1 is selected from the group consisting of ═N— and ═CR7b—;
    • X2 is selected from the group consisting of ═N— and ═CR7c—;
    • X3 is selected from the group consisting of ═N— and ═CR7d—; and
    • R7a, R7b, R7c, and R7d is independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, —OZ1, and —(CH2)q—NR6aR6b;
    • q is 0 or 1.


Embodiment 2. A compound of Formula II:




embedded image


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


R1 is selected from the group consisting of:

    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl,
        • (6) —OZ1,
        • (7) C2-C6 alkenyl, and
        • (8) C2-C6 alkynyl,
      • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1,
      • (c) C2-C6 alkynyl,
    • (iii) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (iv) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl,
      • (f) —OZ1, and
      • (g) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, C1-C6 alkyl, and C1-C6 haloalkyl, and —OZ1,
    • (v) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vi) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vii) —S(═O)2Z2, and
    • (viii) —C(═O)Z2;


Y1 is selected from the group consisting of ═CR2—, ═N—, and —C(═O)—;


with the provisos that:

    • (i) custom-character represents a double bond when Y1 is ═CR2— or ═N—, and R4 is absent; or
    • (ii) custom-character represents a single bond when Y1 is —C(═O)—, and R4 is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R2 is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)n—NR5aR5b;


      n is 0 or 1;


      R5a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) —C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2, and
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4,
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R5b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      R3 and R4 are independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) —(CH2)p—NR6aR6b;


      p is 0 or 1;


      R6a is selected from the group consisting of:
    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) hydroxy,
      • (c) cyano,
      • (d) —OZ1,
      • (e) —SZ1,
      • (f) —NZ3Z4,
      • (g) —C(═O)Z2
      • (h) C(═O)OH,
      • (i) —C(═O)OZ2,
      • (j) —C(═O)NZ3Z4,
      • (k) —NZ5C(═O)Z2,
      • (l) —NZ5C(═O)OZ2,
      • (m) —NZ5C(═O)NZ3Z4,
      • (n) —S(═O)2Z8,
      • (o) —S(═O)2NZ3Z4,
      • (p) —S(═O)(═NZ6)Z2,
      • (q) —S(═Z6)(═NZ7)Z2,
      • (r) —S(═O)(═NZ6)NZ3Z4,
      • (s) —NZ5S(═O)2Z2,
      • (t) —NZ5S(═O)2NZ3Z4,
      • (u) —NZ5S(═O)(═NZ6)Z2,
      • (v) —NZ5S(═NZ6)(═NZ7)Z2, and
      • (w) —NZ5S(═O)(═NZ6)NZ3Z4,
    • (iii) —C(═O)Z2
    • (iv) —C(═O)OZ2,
    • (v) —C(═O)NZ3Z4,
    • (vi) —S(═O)2Z8,
    • (vii) —S(═O)2NZ3Z4,
    • (viii) —S(═O)(═NZ6)Z2,
    • (xi) —S(═Z6)(═NZ7)Z2, and
    • (x) —S(═O)(═NZ6)NZ3Z4;


      R6b is selected from the group consisting of hydrogen and C1-C6 alkyl;


      each Z1 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z2 is independently selected from the group consisting of:
    • (i) C1-C6 alkyl,
    • (ii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) C2-C6 alkynyl,
    • (iv) C3-C6 cycloalkyl,
    • (v) C3-C6 cycloalkenyl, and
    • (vi) C1-C6 haloalkyl;


      each Z3 is independently selected from the group consisting of:
    • (i) hydrogen;
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z4, Z5, Z6 and Z7 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) C3-C6 cycloalkyl;


      each Z8 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) halogen, and
    • (v) hydroxy;
    • X is selected from the group consisting of ═N— and ═CR7a—;
    • X1 is selected from the group consisting of ═N— and ═CR7b—;
    • X2 is selected from the group consisting of ═N— and ═CR7c—;
    • X3 is selected from the group consisting of ═N— and ═CR7d—; and
    • R7a, R7b, R7c, and R7d is independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, —OZ1, and —NZ3Z4.


Embodiment 3. The compound of Embodiment 1 or 2 of Formula III:




embedded image


or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof.


Embodiment 4. The compound of any one of Embodiments 1-3, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R2 is —(CH2)n—NR5aR5b; and


R3 is selected from the group consisting of hydrogen and C1-C6 alkyl.


Embodiment 5. The compound of any one of Embodiments 1-4, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R5a is —C(═O)Z2 and R5b is hydrogen.


Embodiment 6. The compound of any one of Embodiments 1-5, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R2 is selected from the group consisting of hydrogen and C1-C6 alkyl; and


R3 is —(CH2)p—NR6aR6b.


Embodiment 7. The compound of any one of Embodiments 1-6, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R6a is —C(═O)Z2 and R6b is hydrogen.


Embodiment 8. The compound of Embodiment 1 or 2 of Formula IV:




embedded image


or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof.


Embodiment 9. The compound of any one of Embodiments 1, 2, or 8, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R3 is —(CH2)p—NR6aR6b.


Embodiment 10. The compound of any one of Embodiments 1, 2, 8, or 9, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R6a is selected from the group consisting of —C(═O)Z2 and —S(═O)2Z8; and


R6b is hydrogen.


Embodiment 11. The compound of Embodiment 1 or 2 of Formula V:




embedded image


or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof.


Embodiment 12. The compound of any one of Embodiments 1, 2, or 11, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R3 is —(CH2)p—NR6aR6b.


Embodiment 13. The compound of any one of Embodiments 1, 2, 11, or 12, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


R6a is selected from the group consisting of —C(═O)Z2; and


R6b is hydrogen.


Embodiment 14. The compound of any one of Embodiments 1, 2, or 11-13, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R1 is unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, and —OZ1.


Embodiment 15. The compound of any one of Embodiments 1, 2, or 11-14, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein:


X is —CR7a═;


X1 is —CR7b═;


X2 is —CR7c═; and


X3 is —CR7d═.


Embodiment 16. The compound of Embodiment 1, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of:

  • N-[[5-(cyanomethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-[[rac-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-[[(3S)-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-[[(3R)-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-phenoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[6-(cyclopropoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(3-methyl-2,6-dioxo-pyrimidin-4-yl)oxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-benzyloxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[1-[4-(trifluoromethyl)phenyl]pyrazolo[3,4-c]pyridin-3-yl]methyl]prop-2-enamide;
  • N-[[5-prop-2-ynoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(cyclopropylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[5-(cyclopropylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]methanesulfonamide;
  • 2-[[[1-[4-(trifluoromethyl)phenyl]indazol-3-yl]amino]methyl]prop-2-enoic acid;
  • N-[[5-(trifluoromethyl)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;
  • N-[[6-(trifluoromethyl)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide; and
  • N-[[1-(4,4-difluorocyclohexyl)indazol-3-yl]methyl]prop-2-enamide.


Embodiment 17. The compound of Embodiment 1, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of:

  • N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acrylamide;
  • N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acetamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)propionamide;
  • N-(1-phenyl-1H-indol-3-yl)acrylamide;
  • N-(1-(4-isopropylphenyl)-1H-indol-3-yl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)acrylamide;
  • N-(1-(4-chlorophenyl)-1H-indol-3-yl)acrylamide;
  • N-(1-(3-chlorophenyl)-1H-indol-3-yl)acrylamide;
  • N-(1-([1,1′-biphenyl]-4-yl)-1H-indol-3-yl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)propionamide;
  • N-((1-(3-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acrylamide;
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide;
  • N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acrylamide;
  • N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acetamide; and
  • N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)propionamide.


Embodiment 18. A compound of Formula VI:




embedded image


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


R8 is selected from the group consisting of:

    • (i) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl,
        • (6) —OZ1,
        • (7) C2-C6 alkenyl, and
        • (8) C2-C6 alkynyl,
      • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1,
      • (c) C2-C6 alkynyl,
    • (ii) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (iii) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (iv) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (v) unsubstituted or substituted C3-C6 heterocycle, wherein one or more substituents are independently selected from the group consisting of:
      • (a) halogen,
      • (b) cyano,
      • (c) C1-C6 alkyl,
      • (d) C3-C6 cycloalkyl,
      • (e) C1-C6 haloalkyl, and
      • (f) —OZ1,
    • (vi) —S(═O)2Z2, and
    • (vii) —C(═O)Z2;


X is —N(R11)—; or X is absent;


R11 is selected from the group consisting of hydrogen and C1-C6 alkyl;


R9 is selected from the group consisting of:




embedded image


R12 is selected from the group consisting of:

    • (i) hydrogen,
    • (ii) unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of:
      • (a) unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl,
        • (6) —OZ1,
        • (7) C2-C6 alkenyl, and
        • (8) C2-C6 alkynyl,
      • (b) unsubstituted or substituted C3-C6 cycloalkyl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1, and
      • (c) unsubstituted or substituted 5- to 9-membered heteroaryl, wherein one or more substituents are independently selected from the group consisting of:
        • (1) halogen,
        • (2) cyano,
        • (3) C1-C6 alkyl,
        • (4) C3-C6 cycloalkyl,
        • (5) C1-C6 haloalkyl, and
        • (6) —OZ1, and
    • (iii) C1-C6 haloalkyl;


R10 is —(CH2)q—C(═O)R13


q is 0, 1, or 2;


R13 is selected from the group consisting of —OH, OZ1, and —NZ3Z4;


each Z1 is independently selected from the group consisting of:

    • (i) hydrogen,
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl;


      each Z2 is independently selected from the group consisting of:
    • (i) C1-C6 alkyl,
    • (ii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iii) C2-C6 alkynyl,
    • (iv) C3-C6 cycloalkyl,
    • (v) C3-C6 cycloalkenyl, and
    • (vi) C1-C6 haloalkyl;


      each Z3 is independently selected from the group consisting of:
    • (i) hydrogen;
    • (ii) C1-C6 alkyl,
    • (iii) unsubstituted or substituted C2-C6 alkenyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, halogen and unsubstituted or substituted C1-C6 alkyl, wherein one or more substituents are independently selected from the group consisting of cyano, —S(═O)2Z8, —NZ3Z4 and 4- to 8-membered heterocycle,
    • (iv) C2-C6 alkynyl,
    • (v) C3-C6 cycloalkyl,
    • (vi) C3-C6 cycloalkenyl, and
    • (vii) C1-C6 haloalkyl; and


      each Z4 is independently selected from the group consisting of:
    • (i) hydrogen,
    • (ii) C1-C6 alkyl, and
    • (iii) C3-C6 cycloalkyl.


Embodiment 19. The compound of Embodiment 18, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of:

  • 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetic acid;
  • N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetamide;
  • 2-(6-phenyl-8-(1-(pyridin-3-ylmethyl)-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-2-yl)acetic acid;
  • 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetic acid; and
  • N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetamide.


Embodiment 20. A compound of Table 1.


Embodiment 21. A pharmaceutical composition comprising the compound of any one of Embodiments 1-20, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, and a pharmaceutically acceptable carrier.


Embodiment 22. The compound of any one of Embodiments 1-20, or the pharmaceutical composition of claim 21, for use as a medicine.


Embodiment 23. The compound of any one of Embodiments 1-20, or the pharmaceutical composition of Embodiment 21, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorder in an animal, mammal or human.


Embodiment 24. The compound of any one of Embodiments 1-20, or the pharmaceutical composition of Embodiment 21, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorder selected from the group comprising cancer, fibrosis and YAP/TAZ-TEAD activation mediated congenital disorders.


Embodiment 25. The compound of any one of Embodiments 1-20, or the pharmaceutical composition of Embodiment 21, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorder selected from lung cancer, breast cancer, head and neck cancer, oesophageal cancer, kidney cancer, bladder cancer, colon cancer, ovarian cancer, cervical cancer, endometrial cancer, liver cancer (including but not limited to cholangiocarcinoma), skin cancer, pancreatic cancer, gastric cancer, brain cancer and prostate cancer, mesotheliomas, and/or sarcomas.


Embodiment 26. The compound of any one of Embodiments 1-20, or the pharmaceutical composition of Embodiment 21, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorder selected from acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute T-cell leukemia, basal cell carcinoma, bile duct carcinoma, bronchogenic carcinoma, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, glioblastoma, gliosarcoma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, leukemia, liposarcoma, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, NUT midline carcinoma (NMC), non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenstrom's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor.


Embodiment 27. A method for the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorders in an animal, mammal or human comprising administering to said animal, mammal or human in need for such prevention or treatment an effective dose of the compounds of any one of Embodiments 1-20.


Embodiment 28. A method of treatment or prevention of YAP/TAZ-TEAD activation mediated disorder according to Embodiment 27 to a patient in need thereof in combination with one or more other medicines selected from the group consisting of EGFR inhibitors, MEK inhibitors, AXL inhibitors, B-RAF inhibitors, and RAS inhibitors.


More generally, the disclosure relates to the compounds of the formulae described herein and embodiments, statements and aspects thereof being useful as agents having biological activity or as diagnostic agents. Any of the uses mentioned with respect to the present disclosure may be restricted to a non-medical use, a non-therapeutic use, a non-diagnostic use, or exclusively an in vitro use, or a use related to cells remote from an animal.


Compounds of the present disclosure are small molecule YAP/TAZ-TEAD inhibitors. Small molecule YAP/TAZ-TEAD inhibitors are useful, e.g., for the treatment of cancer, including with no limitations, lung cancer, breast cancer, head and neck cancer, oesophageal cancer, kidney cancer, bladder cancer, colon cancer, ovarian cancer, cervical cancer, endometrial cancer, liver cancer (including but not limited to cholangiocarcinoma), skin cancer, pancreatic cancer, gastric cancer, brain cancer and prostate cancer, mesotheliomas, and/or sarcomas. In other embodiments, small molecule YAP/TAZ-TEAD inhibitors are useful for the treatment of cancers characterized by squamous cell carcinomas of the lung, cervix, ovaries, head and neck, oesophagus, and/or skin. In other embodiments, small molecule YAP/TAZ-TEAD inhibitors are useful for the treatment of cancers that originate from neuroectoderm-derived tissues, such as ependymomas, meningiomas, schwannomas, peripheral nerve-sheet tumors and/or neuroblastomas. In other embodiments, small molecule YAP/TAZ-TEAD inhibitors are useful for the treatment of vascular cancers, such as epithelioid haemangioendotheliomas, or for the treatment of supratentorial ependymomas or porocarcinomas. In some embodiments, the solid tumors have gain-of-function gene amplifications, gene fusions or activating mutations in the YAP1 or WWTR1 (TAZ) genes. In some embodiments the solid tumors have loss-of-function mutations or deletions in the NF2, LATS1/2, BAP1, FAT1, SAV1, and/or MST1/2 genes. In some embodiments solid tumors have gain-of-function mutations in the GNAQ and/or GNA11 genes, e.g. in uveal melanoma. In some embodiments, solid cancer are characterized by constitutive nuclear presence of YAP and/or TAZ. In some embodiments, solid cancers are characterized by the overexpression of YAP/TAZ-TEAD signature genes, including but not limited to CTGF, CYR61, AMOTL2, and/or ANKRD1.


Small molecule YAP/TAZ-TEAD inhibitors may also be useful to treat cancers that have developed resistance to prior treatments. This may include, for instance, the treatment of cancers that have developed resistance to chemotherapy, or to targeted therapy. In some embodiments, this may include the treatment of cancers that have developed resistance to inhibitors of receptor tyrosine kinases, such as EGFR (afatinib, erlotinib hydrochloride, osimertinib, gefitinib, dacomitinib, neratinib, canertinib, cetuximab) or AXL (crizotinib, cabozantinib, gilteritinib, sitravatinib, bemcentinib, dubermatinib), to components of the RAS-MAPK signaling cascade, including inhibitors of RAS itself (such as AMG510, MRTX849, B11701963, ARS1620), inhibitors of B-RAF (sorafinib tosylate, dabrafenib, vemurafenib, regorafenib), or MEK1/2 (trametinib, selumetinib, cobimetinib, mirdametinib).


Small molecule YAP/TAZ-TEAD inhibitors may also be useful when combined, upon simultaneous administration, or subsequent administration, with other agents used for the treatment of cancer. This may include, for instance, the co-treatment with inhibitors or monoclonal antibodies targeting receptor tyrosine kinases such as EGFR (afatinib, erlotinib hydrochloride, osimertinib, gefitinib, dacomitinib, neratinib, canertinib, cetuximab) or AXL (crizotinib, cabozantinib, gilteritinib, sitravatinib, bemcentinib, dubermatinib), to components of the RAS-MAPK signaling cascade, including inhibitors of RAS itself (such as AMG510, MRTX849, BI1701963, ARS1620), inhibitors of B-RAF (sorafinib tosylate, dabrafenib, vemurafenib, regorafenib), or MEK1/2 (trametinib, selumetinib, cobimetinib, mirdametinib).


Small molecule YAP/TAZ-TEAD inhibitors may also be useful to treat a metastasized cancer. In some instances, the metastasized cancer is selected from metastasized uveal melanoma, mesothelioma, esophageal cancer, liver cancer, breast cancer, hepatocellular carcinoma, lung adenocarcinoma, glioma, colon cancer, colorectal cancer, gastric cancer, medulloblastoma, ovarian cancer, esophageal squamous cell carcinoma, sarcoma, Ewing sarcoma, head and neck cancer, prostate cancer, and meningioma.


In some embodiments, the cancer treated could be malignant pleural mesothelioma or lung cancer.


In some embodiments, the compounds of the disclosure can be used for the treatment of acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute T-cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, glioblastoma, gliosarcoma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, leukemia, liposarcoma, lung cancer, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, NUT midline carcinoma (NMC), non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenstrom's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor.


Malignant Pleural Mesothelioma


Small molecule YAP/TAZ-TEAD inhibitors may also be useful to treat malignant pleural mesothelioma, as a single agent, or in combination with inhibitors such as pemetrexed disodium, raltitrexed, carboplatin, oxaliplatin, gemcitabine, doxorubicin, or monoclonal antibodies such as bevacizumab. Combinations with checkpoint inhibitors such as pembrolizumab, atezolizumab, and/or nivolumab. Combinations with cell therapy, for instance, chimeric antigen receptor (CAR) T therapy or CAR NK therapy, which may, for instance, use mesothelin (MSLN) as an antigen. Combinations with monoclonal antibodies that, for instance, recognize mesothelin as an antigen, for instance BMS-986148, BAY 94-9343, amatuximab, and/or LMB-100.


Lung Cancer


Small molecule YAP/TAZ-TEAD inhibitors may also be useful to treat lung cancer, as a single agent, or in combination with inhibitors such as afatinib, bevacizumab, cabozantinib, ceritinib, crizotinib, erlotinib hydrochloride, osimertinib, ramucirumab, gefitinib, alectinib, trastuzumab, cetuximab, ipilimumab, trametinib, dabrafenib, vemurafenib, dacomitinib, tivantinib, and/or onartuzumab. Combinations with checkpoint inhibitors such as pembrolizumab, atezolizumab, and/or nivolumab. Combinations with cisplatin, carboplatin, paclitaxel, paclitaxel protein bound, docetaxel, gemcitabine, vinorelbine, etoposide, nintedanib, vinblastine, pemetrexed, afatinib, bevacizumab, cabozantinib, ceritinib, crizotinib, erlotinib hydrochloride, osimertinib, ramucirumab, gefitinib, necitumumab, alectinib, trastuzumab, cetuximab, ipilimumab, trametinib, dabrafenib, vemurafenib, dacomitinib, tivantinib, onartuzumab, pembrolizumab, atezolizumab, and/or nivolumab


In some embodiments, small molecule YAP/TAZ-TEAD inhibitors are useful, e.g., for the treatment of congenital disorders. In some embodiments, the congenital disease is mediated by activation of transcriptional coactivator with PDZ binding motif/Yes-associated protein transcription coactivator (TAZ/YAP). In some embodiments, the congenital disease is characterized by a mutant Ga-protein. In some embodiments, the mutant Ga-protein is selected from G12, G13, Gq, G11, Gi, Go, and Gs. In some embodiments, the congenital disease is characterized by loss-of-function mutations or deletions in the NF2 gene. Exemplary congenital diseases include, but are not limited to, Sturge-Weber Syndrome, Port-Wine stain, and Neurofibromatosis. In some embodiments the congenital disease is Neurofibromatosis, including but not limited to Neurofibromatosis type 2.


In some embodiments, small molecule YAP/TAZ-TEAD inhibitors are useful, e.g., for the treatment of fibrotic disorders, such as fibrosis of the liver, the lung, the kidney, the heart or the skin. In some embodiments, fibrosis can be treated in the context of non-alcoholic fatty liver disease, primary sclerosing cholangitis, primary biliary cirrhosis, idiopathic pulmonary fibrosis, chronic kidney disease, and/or myocardial infarction injury.


The compounds of the disclosure can inhibit YAP/TAZ-TEAD transcription activation. The compounds have been shown to inhibit YAP/TAZ-TEAD transcription activity in cellular models and in an animal model. The compounds have also been shown to have an inhibitory effect on cancer cell lines that are dependent on YAP/TAZ-TEAD transcription activity and on the growth of cancer in a xenograft cancer model.


The compounds of the disclosure can optionally be bound covalently to an insoluble matrix and used for affinity chromatography (separations, depending on the nature of the groups of the compounds, for example compounds with pendant aryl are useful in hydrophobic affinity separations).


When using one or more derivatives of the formulae as defined herein:

    • the active ingredients of the compound(s) may be administered to the animal or mammal (including a human) to be treated by any means well known in the art, i.e. orally, intranasally, subcutaneously, intramuscularly, intradermally, intravenously, intra-arterially, parenterally or by catheterization.
    • the therapeutically effective amount of the preparation of the compound(s), especially for the treatment of diseases mediated by activity of YAP/TAZ-TEAD transcription in humans and other mammals (such as cancer, fibrosis and certain congenital disorders), preferably is a YAP/TAZ-TEAD transcription inhibiting amount of the compounds of the formulae, statements, aspects and embodiments as defined herein and corresponds to an amount which ensures a plasma level that is able to inhibit the YAP/TAZ-TEAD activation and is between 1 μg/ml and 100 mg/ml.


Suitable dosages of the compounds or compositions of the disclosure should be used to treat or prevent the targeted diseases in a subject. Depending upon the pathologic condition to be treated and the patient's condition, the said effective amount may be divided into several sub-units per day or may be administered at more than one day intervals.


According to a particular embodiment of the disclosure, the compounds of the disclosure may be employed in combination with other therapeutic agents for the treatment or prophylaxis of diseases mediated by activity of YAP/TAZ-TEAD transcription in humans and other mammals (such as cancer, fibrosis and certain congenital disorders). The disclosure therefore relates to the use of a composition comprising:

  • (a) one or more compounds of the formulae and aspects, statements and embodiments herein, and
  • (b) one or more further therapeutic or preventive agents that are used for the prevention or treatment of cancer or fibrosis as biologically active agents in the form of a combined preparation for simultaneous, separate or sequential use.


    The compound or composition can be administered concurrently with, prior to, or subsequent to the one or more additional therapeutic agents, which are different from the compound described herein and may be useful as, e.g., combination therapies.


Examples of such further therapeutic agents for use in combinations include agents that are inhibitors of:

    • EGFR (such as afatinib, erlotinib hydrochloride, osimertinib, gefitinib, dacomitinib, neratinib, canertinib, cetuximab),
    • AXL (such as crizotinib, cabozantinib, gilteritinib, sitravatinib, bemcentinib, dubermatinib),
    • components of the RAS-MAPK signaling cascade, including inhibitors of RAS itself (such as AMG510, MRTX849, B11701963, ARS1620),
    • B-RAF (such as sorafinib tosylate, dabrafenib, vemurafenib, regorafenib), or
    • MEK1/2 (trametinib, selumetinib, cobimetinib, mirdametinib).


The pharmaceutical composition or combined preparation according to this disclosure may contain the compounds of the present disclosure over a broad content range depending on the contemplated use and the expected effect of the preparation. Generally, the content of the derivatives of the present disclosure of the combined preparation is within the range of 0.1 to 99.9% by weight, preferably from 1 to 99% by weight, more preferably from 5 to 95% by weight.


Those of skill in the art will also recognize that the compounds of the disclosure may exist in many different protonation states, depending on, among other things, the pH of their environment. While the structural formulae provided herein depict the compounds in only one of several possible protonation states, it will be understood that these structures are illustrative only, and that the disclosure is not limited to any particular protonation state—any and all protonated forms of the compounds are intended to fall within the scope of the disclosure.


The term “pharmaceutically acceptable salts” as used herein means the therapeutically active non-toxic salt forms which the compounds of formulae herein are able to form. Therefore, the compounds of this disclosure optionally comprise salts of the compounds herein, especially pharmaceutically acceptable non-toxic salts containing, for example, Na+, Li+, K+, Ca2+ and Mg2+. Such salts may include those derived by combination of appropriate cations such as alkali and alkaline earth metal ions or ammonium and quaternary amino ions with an acid anion moiety, typically a carboxylic acid. The compounds of the disclosure may bear multiple positive or negative charges. The net charge of the compounds of the disclosure may be either positive or negative. Any associated counter ions are typically dictated by the synthesis and/or isolation methods by which the compounds are obtained. Typical counter ions include, but are not limited to ammonium, sodium, potassium, lithium, halides, acetate, trifluoroacetate, etc., and mixtures thereof. It will be understood that the identity of any associated counter ion is not a critical feature of the disclosure, and that the disclosure encompasses the compounds in association with any type of counter ion. Moreover, as the compounds can exist in a variety of different forms, the disclosure is intended to encompass not only forms of the compounds that are in association with counter ions (e.g., dry salts), but also forms that are not in association with counter ions (e.g., aqueous or organic solutions). Metal salts typically are prepared by reacting the metal hydroxide with a compound of this disclosure. Examples of metal salts which are prepared in this way are salts containing Li+, Na+, and K+. A less soluble metal salt can be precipitated from the solution of a more soluble salt by addition of the suitable metal compound. In addition, salts may be formed from acid addition of certain organic and inorganic acids to basic centers, typically amines, or to acidic groups. Examples of such appropriate acids include, for instance, inorganic acids such as hydrohalogen acids, e.g. hydrochloric or hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, 2-hydroxypropanoic, 2-oxopropanoic, lactic, pyruvic, oxalic (i.e. ethanedioic), malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclohexanesulfamic, salicylic (i.e. 2-hydroxybenzoic), p-aminosalicylic and the like. Furthermore, this term also includes the solvates which the compounds of formulae herein as well as their salts are able to form, such as for example hydrates, alcoholates and the like. Finally, it is to be understood that the compositions herein comprise compounds of the disclosure in their unionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.


Also included within the scope of this disclosure are the salts of the parental compounds with one or more amino acids, especially the naturally-occurring amino acids found as protein components. The amino acid typically is one bearing a side chain with a basic or acidic group, e.g., lysine, arginine or glutamic acid, or a neutral group such as glycine, serine, threonine, alanine, isoleucine, or leucine.


The compounds of the disclosure also include physiologically acceptable salts thereof. Examples of physiologically acceptable salts of the compounds of the disclosure include salts derived from an appropriate base, such as an alkali metal (for example, sodium), an alkaline earth (for example, magnesium), ammonium and NX4+ (wherein X is C1-C4 alkyl). Physiologically acceptable salts of an hydrogen atom or an amino group include salts of organic carboxylic acids such as acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, sulfuric, phosphoric and sulfamic acids. Physiologically acceptable salts of a compound containing a hydroxy group include the anion of said compound in combination with a suitable cation such as Na+ and NX4+ (wherein X typically is independently selected from H or a C1-C4 alkyl group). However, salts of acids or bases which are not physiologically acceptable may also find use, for example, in the preparation or purification of a physiologically acceptable compound. All salts, whether or not derived form a physiologically acceptable acid or base, are within the scope of the present disclosure.


As used herein and unless otherwise stated, the term “enantiomer” means each individual optically active form of a compound of the disclosure, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (e.g. at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.


The term “isomers” as used herein means all possible isomeric forms, including tautomeric and stereochemical forms, which the compounds of formulae herein may possess, but not including position isomers. Typically, the structures shown herein exemplify only one tautomeric or resonance form of the compounds, but the corresponding alternative configurations are contemplated as well. Unless otherwise stated, the chemical designation of compounds denotes the mixture of all possible stereochemically isomeric forms, said mixtures containing all diastereomers and enantiomers (since the compounds of formulae herein may have at least one chiral center) of the basic molecular structure, as well as the stereochemically pure or enriched compounds. More particularly, stereogenic centers may have either the R- or S-configuration, and multiple bonds may have either cis- or trans-configuration.


Pure isomeric forms of the said compounds are defined as isomers substantially free of other enantiomeric or diastereomeric forms of the same basic molecular structure. In particular, the term “stereoisomerically pure” or “chirally pure” relates to compounds having a stereoisomeric excess of at least about 80% (e.g. at least 90% of one isomer and at most 10% of the other possible isomers), preferably at least 90%, more preferably at least 94% and most preferably at least 97%. The terms “enantiomerically pure” and “diastereomerically pure” should be understood in a similar way, having regard to the enantiomeric excess, respectively the diastereomeric excess, of the mixture in question.


Separation of stereoisomers is accomplished by standard methods known to those in the art. One enantiomer of a compound of the disclosure can be separated substantially free of its opposing enantiomer by a method such as formation of diastereomers using optically active resolving agents (“Stereochemistry of Carbon Compounds,” (1962) by E. L. Eliel, McGraw Hill; Lochmuller, C. H., (1975) J. Chromatogr., 113:(3) 283-302). Separation of isomers in a mixture can be accomplished by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure enantiomers, or (3) enantiomers can be separated directly under chiral conditions. Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, α-methyl-b-phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts. Alternatively, by method (2), the substrate to be resolved may be reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. (1994) Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the free, enantiomerically enriched compound. A method of determining optical purity involves making chiral esters, such as a menthyl ester or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob III. (1982) J. Org. Chem. 47:4165), of the racemic mixture, and analyzing the NMR spectrum for the presence of the two atropisomeric diastereomers. Stable diastereomers can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (Hoye, T., WO 96/15111). Under method (3), a racemic mixture of two asymmetric enantiomers is separated by chromatography using a chiral stationary phase. Suitable chiral stationary phases are, for example, polysaccharides, in particular cellulose or amylose derivatives. Commercially available polysaccharide based chiral stationary phases are ChiralCel™ CA, OA, OB5, OC5, OD, OF, OG, OJ and OK, and Chiralpak™ AD, AS, OP(+) and OT(+). Appropriate eluents or mobile phases for use in combination with said polysaccharide chiral stationary phases are hexane and the like, modified with an alcohol such as ethanol, isopropanol and the like. (“Chiral Liquid Chromatography” (1989) W. J. Lough, Ed. Chapman and Hall, New York; Okamoto, (1990) “Optical resolution of dihydropyridine enantiomers by High-performance liquid chromatography using phenylcarbamates of polysaccharides as a chiral stationary phase”, J. of Chromatogr. 513:375-378).


The terms cis and trans are used herein in accordance with Chemical Abstracts nomenclature and include reference to the position of the substituents on a ring moiety. The absolute stereochemical configuration of the compounds of the formulae described herein may easily be determined by those skilled in the art while using well-known methods such as, for example, X-ray diffraction.


The present disclosure also includes isotopically labelled compounds, which are identical to those recited in the formulas recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that may be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2H, 3H, 13C, 11C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, and 36Cl, respectively. Compounds of the present disclosure and pharmaceutically acceptable salts of said compounds or which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this disclosure. Certain isotopically labeled compounds of the present disclosure, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labelled compounds of the formulas of this disclosure may generally be prepared by carrying out the procedures disclosed in the examples and preparations described herein, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.


Also encompassed within the disclosure are modifications of the compounds of formula (I) or other formulas, embodiments, aspects or parts thereof or metabolites thereof using PROTAC technology (Schapira M. et al, Nat. Rev. Drug Discov. 2019, 18(12), 949-963). Specifically, the PROTAC technology designs a bifunctional small molecule, one end of which is a compound of the general formula (I) or other formulas, embodiments, aspects or parts thereof or metabolites thereof, and the other end of which is connected with a ligand of E3 ubiquitin ligase through a connecting chain, to form a target-induced protein degradation complex. Because this degradation has a catalytic effect, a lower dosage can achieve efficient degradation. The compound of the general formula (I) or other formulas, embodiments, aspects or parts thereof or metabolites thereof can be connected via a linker arm (e.g. long-chain ethylene glycol with the length of 2-10, long-chain propylene glycol with the length of 2-10 and long-chain fatty alkane with the length of 2-10) to a ligand of E3 ubiquitin ligase such as e.g. thalidomide analogs.


The compounds of the disclosure may be formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. Formulations optionally contain excipients such as those set forth in the “Handbook of Pharmaceutical Excipients” (1986) and include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like.


Subsequently, the term “pharmaceutically acceptable carrier” as used herein means any material or substance with which the active ingredient is formulated in order to facilitate its application or dissemination to the locus to be treated, for instance by dissolving, dispersing or diffusing the said composition, and/or to facilitate its storage, transport or handling without impairing its effectiveness. The pharmaceutically acceptable carrier may be a solid or a liquid or a gas which has been compressed to form a liquid, e.g. the compositions of this disclosure can suitably be used as concentrates, emulsions, solutions, granulates, dusts, sprays, aerosols, suspensions, ointments, creams, tablets, pellets or powders.


Suitable pharmaceutical carriers for use in the said pharmaceutical compositions and their formulation are well known to those skilled in the art, and there is no particular restriction to their selection within the present disclosure. They may also include additives such as wetting agents, dispersing agents, stickers, adhesives, emulsifying agents, solvents, coatings, antibacterial and antifungal agents (for example phenol, sorbic acid, chlorobutanol), isotonic agents (such as sugars or sodium chloride) and the like, provided the same are consistent with pharmaceutical practice, e.g. carriers and additives which do not create permanent damage to mammals. The pharmaceutical compositions of the present disclosure may be prepared in any known manner, for instance by homogeneously mixing, coating and/or grinding the active ingredients, in a one-step or multi-steps procedure, with the selected carrier material and, where appropriate, the other additives such as surface-active agents. may also be prepared by micronisation, for instance in view to obtain them in the form of microspheres usually having a diameter of about 1 to 10 gm, namely for the manufacture of microcapsules for controlled or sustained release of the active ingredients.


Suitable surface-active agents, also known as emulgent or emulsifier, to be used in the pharmaceutical compositions of the present disclosure are non-ionic, cationic and/or anionic materials having good emulsifying, dispersing and/or wetting properties. Suitable anionic surfactants include both water-soluble soaps and water-soluble synthetic surface-active agents. Suitable soaps are alkaline or alkaline-earth metal salts, unsubstituted or substituted ammonium salts of higher fatty acids (C10-C22), e.g. the sodium or potassium salts of oleic or stearic acid, or of natural fatty acid mixtures obtainable from coconut oil or tallow oil. Synthetic surfactants include sodium or calcium salts of polyacrylic acids; fatty sulphonates and sulphates; sulphonated benzimidazole derivatives and alkylarylsulphonates. Fatty sulphonates or sulphates are usually in the form of alkaline or alkaline-earth metal salts, unsubstituted ammonium salts or ammonium salts substituted with an alkyl or acyl group having from 8 to 22 carbon atoms, e.g. the sodium or calcium salt of lignosulphonic acid or dodecylsulphonic acid or a mixture of fatty alcohol sulphates obtained from natural fatty acids, alkaline or alkaline-earth metal salts of sulphuric or sulphonic acid esters (such as sodium lauryl sulphate) and sulphonic acids of fatty alcohol/ethylene oxide adducts. Suitable sulphonated benzimidazole derivatives preferably contain 8 to 22 carbon atoms. Examples of alkylarylsulphonates are the sodium, calcium or alcoholamine salts of dodecylbenzene sulphonic acid or dibutyl-naphthalenesulphonic acid or a naphthalene-sulphonic acid/formaldehyde condensation product. Also suitable are the corresponding phosphates, e.g. salts of phosphoric acid ester and an adduct of p-nonylphenol with ethylene and/or propylene oxide, or phospholipids. Suitable phospholipids for this purpose are the natural (originating from animal or plant cells) or synthetic phospholipids of the cephalin or lecithin type such as e.g. phosphatidylethanolamine, phosphatidylserine, phosphatidylglycerine, lysolecithin, cardiolipin, dioctanylphosphatidyl-choline, dipalmitoylphoshatidyl-choline and their mixtures.


Suitable non-ionic surfactants include polyethoxylated and polypropoxylated derivatives of alkylphenols, fatty alcohols, fatty acids, aliphatic amines or amides containing at least 12 carbon atoms in the molecule, alkylarenesulphonates and dialkylsulphosuccinates, such as polyglycol ether derivatives of aliphatic and cycloaliphatic alcohols, saturated and unsaturated fatty acids and alkylphenols, said derivatives preferably containing 3 to 10 glycol ether groups and 8 to 20 carbon atoms in the (aliphatic) hydrocarbon moiety and 6 to 18 carbon atoms in the alkyl moiety of the alkylphenol. Further suitable non-ionic surfactants are water-soluble adducts of polyethylene oxide with polypropylene glycol, ethylenediaminopolypropylene glycol containing 1 to 10 carbon atoms in the alkyl chain, which adducts contain 20 to 250 ethyleneglycol ether groups and/or 10 to 100 propyleneglycol ether groups. Such compounds usually contain from 1 to 5 ethyleneglycol units per propyleneglycol unit. Representative examples of non-ionic surfactants are nonylphenol-polyethoxyethanol, castor oil polyglycolic ethers, polypropylene/polyethylene oxide adducts, tributylphenoxypolyethoxyethanol, polyethyleneglycol and octylphenoxypolyethoxyethanol. Fatty acid esters of polyethylene sorbitan (such as polyoxyethylene sorbitan trioleate), glycerol, sorbitan, sucrose and pentaerythritol are also suitable non-ionic surfactants.


Suitable cationic surfactants include quaternary ammonium salts, particularly halides, having 4 hydrocarbon groups optionally substituted with halo, phenyl, substituted phenyl or hydroxy; for instance quaternary ammonium salts containing as N-substituent at least one C8-22alkyl (e.g. cetyl, lauryl, palmityl, myristyl, oleyl and the like) and, as further substituents, unsubstituted or halogenated lower alkyl, benzyl and/or hydroxy-lower alkyl.


A more detailed description of surface-active agents suitable for this purpose may be found for instance in “McCutcheon's Detergents and Emulsifiers Annual” (MC Publishing Crop., Ridgewood, N.J., 1981), “Tensid-Taschenbucw’, 2 d ed. (Hanser Verlag, Vienna, 1981) and “Encyclopaedia of Surfactants, (Chemical Publishing Co., New York, 1981).


Compounds of the disclosure and their pharmaceutically acceptable salts (hereafter collectively referred to as the active ingredients) may be administered by any route appropriate to the condition to be treated, suitable routes including oral, rectal, nasal, topical (including ocular, buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural). The preferred route of administration may vary with for example the condition of the recipient.


While it is possible for the active ingredients to be administered alone it is preferable to present them as pharmaceutical formulations. The formulations, both for veterinary and for human use, of the present disclosure comprise at least one active ingredient, as above described, together with one or more pharmaceutically acceptable carriers therefore and optionally other therapeutic ingredients. The carrier(s) optimally are “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. The formulations include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural) administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.


Formulations of the present disclosure suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.


A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. For infections of the eye or other external tissues e.g. mouth and skin, the formulations are optionally applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1% and 20% in increments of 0.1% w/w such as 0.6% w/w, 0.7% w/w, etc), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w. When formulated in an ointment, the active ingredients may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase of the cream base may include, for example, at least 30% w/w of a polyhydric alcohol, e.g. an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethylsulfoxide and related analogs.


The oily phase of the emulsions of this disclosure may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Optionally, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.


The choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties, since the solubility of the active compound in most oils likely to be used in pharmaceutical emulsion formulations is very low. Thus the cream should optionally be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.


Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is optionally present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w. Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.


Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate. Formulations suitable for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns (including particle sizes in a range between 20 and 500 microns in increments of 5 microns such as 30 microns, 35 microns, etc), which is administered in the manner in which snuff is taken, e.g. by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable formulations wherein the carrier is a liquid, for administration as for example a nasal spray or as nasal drops, include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol administration may be prepared according to conventional methods and may be delivered with other therapeutic agents.


Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.


Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.


Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient.


It should be understood that in addition to the ingredients particularly mentioned above the formulations of this disclosure may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.


Compounds of the disclosure can be used to provide controlled release pharmaceutical formulations containing as active ingredient one or more compounds of the disclosure (“controlled release formulations”) in which the release of the active ingredient can be controlled and regulated to allow less frequency dosing or to improve the pharmacokinetic or toxicity profile of a given disclosed compound. Controlled release formulations adapted for oral administration in which discrete units comprising one or more compounds of the disclosure can be prepared according to conventional methods.


Additional ingredients may be included in order to control the duration of action of the active ingredient in the composition. Control release compositions may thus be achieved by selecting appropriate polymer carriers such as for example polyesters, polyamino acids, polyvinyl pyrrolidone, ethylene-vinyl acetate copolymers, methylcellulose, carboxymethylcellulose, protamine sulfate and the like. The rate of drug release and duration of action may also be controlled by incorporating the active ingredient into particles, e.g. microcapsules, of a polymeric substance such as hydrogels, polylactic acid, hydroxymethylcellulose, polymethyl methacrylate and the other above-described polymers. Such methods include colloid drug delivery systems like liposomes, microspheres, microemulsions, nanoparticles, nanocapsules and so on. Depending on the route of administration, the pharmaceutical composition may require protective coatings. Pharmaceutical forms suitable for injectionable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation thereof. Typical carriers for this purpose therefore include biocompatible aqueous buffers, ethanol, glycerol, propylene glycol, polyethylene glycol and the like and mixtures thereof.


In view of the fact that, when several active ingredients are used in combination, they do not necessarily bring out their joint therapeutic effect directly at the same time in the mammal to be treated, the corresponding composition may also be in the form of a medical kit or package containing the two ingredients in separate but adjacent repositories or compartments. In the latter context, each active ingredient may therefore be formulated in a way suitable for an administration route different from that of the other ingredient, e.g. one of them may be in the form of an oral or parenteral formulation whereas the other is in the form of an ampoule for intravenous injection or an aerosol.


Another embodiment of this disclosure relates to various precursor or “pro-drug” forms of the compounds of the present disclosure. It may be desirable to formulate the compounds of the present disclosure in the form of a chemical species which itself is not significantly biologically-active, but which when delivered to the animal, mammal or human will undergo a chemical reaction catalyzed by the normal function of the body of the fish, inter alia, enzymes present in the stomach or in blood serum, said chemical reaction having the effect of releasing a compound as defined herein. The term “pro-drug” thus relates to these species which are converted in vivo into the active pharmaceutical ingredient.


The pro-drugs of the compounds of the present disclosure can have any form suitable to the formulator, for example, esters are non-limiting common pro-drug forms. In the present case, however, the pro-drug may necessarily exist in a form wherein a covalent bond is cleaved by the action of an enzyme present at the target locus. For example, a C—C covalent bond may be selectively cleaved by one or more enzymes at said target locus and, therefore, a pro-drug in a form other than an easily hydrolysable precursor, inter alia an ester, an amide, and the like, may be used. The counterpart of the active pharmaceutical ingredient in the pro-drug can have different structures such as an amino acid or peptide structure, alkyl chains, sugar moieties and others as known in the art.


For the purpose of the present disclosure the term “therapeutically suitable pro-drug” is defined herein as “a compound modified in such a way as to be transformed in vivo to the therapeutically active form, whether by way of a single or by multiple biological transformations, when in contact with the tissues of the animal, mammal or human to which the pro-drug has been administered, and without undue toxicity, irritation, or allergic response, and achieving the intended therapeutic outcome”.


More specifically the term “prodrug”, as used herein, relates to an inactive or significantly less active derivative of a compound such as represented by the structural formulae herein described, which undergoes spontaneous or enzymatic transformation within the body in order to release the pharmacologically active form of the compound. For a comprehensive review, reference is made to Rautio J. et al. (“Prodrugs: design and clinical applications” Nature Reviews


Drug Discovery, 2008, doi: 10.1038/nrd2468).


The compounds of the disclosure can be prepared while using a series of chemical reactions well known to those skilled in the art, altogether making up the process for preparing said compounds and exemplified further. The processes described further are only meant as examples and by no means are meant to limit the scope of the present disclosure.


EXAMPLES
General Syntheses

Abbreviations that may be used in the instant specification, particularly in the schemes and examples, are as follows: AIBN—Azobisisobutyronitrile, aq—Aqueous solution, ACN—acetonitrile, Ac2O—Acetic anhydride, AcOH—Acetic acid, BF3·OEt2—Boron trifluoride diethyl etherate, BINAP—(2,2′-bis(diphenylphosphino)-1,1′-binaphthyl), Boc—tert-butyloxycarbonyl, Boc2O—Di-tert-butyl dicarbonate, BrettPhos—2-(Dicyclohexylphosphino)3,6-dimethoxy-2′,4′,6′-triisopropyl-1,1-biphenyl, BrettPhos Pd G3—[(2-Di-cyclohexylphosphino-3,6-dimethoxy-2′,4′,6′-triisopropyl-1,1′-biphenyl)-2-(2′-amino-1,1′-biphenyl)]palladium(II) methanesulfonate, tBuOH—tert-Butanol, Conc—Concentrated, mCPBA—meta-Chloroperoxybenzoic acid, CMBP—(Tributylphosphoranylidene)acetonitrile, Cu(OAc)2—Copper(I1)acetate, d—day, DCM—Dichloromethane, DCE—1,2-Dichloroethane, DIPEA—Diisopropyl ethyl amine, DMA—N,N-Dimethylacetamide, DMAP—N,N-Dimethylpyridin-4-amine, DMF—N,N-Dimethylformamide, DMF-DMA—N,N-Dimethylformamide dimethyl acetal, DMSO—Dimethyl sulfoxide, DMSO-d6-Deuterated dimethyl sulfoxide, DPPA—Diphenyl phosphoryl azide, EDA—Ethyl diazoacetate, EDC—1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide, EtOH—Ethanol, EtOAc—Ethyl acetate, Et2O—Diethyl ether, Eq. —Equivalent, FA—Formic acid, h—Hour, HATU—O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate, HCl—Hydrogen chloride, HNO3—Nitric acid, H2O2—Hydrogen peroxide, HO(CH2O)nH—paraformaldehyde, HOAt—1-Hydroxy-7-azabenzotriazole, HPLC—High performance liquid chromatography, IPA—2-propanol, LAH—Lithiumaluminiumhydride, LG—Leaving group, MeOH—methanol, min—Minute, NBS—N-Bromosuccinimide, Pd(amphos)Cl2 Bis(di-tert-butyl(4-dimethylaminophenyl)phosphine)dichloropalladium(II), Pd(PPh3)2Cl2—Bis(triphenylphosphine)palladium(II)dichloride, Pd(PPh3)4—Tetrakis(triphenylphosphine)palladium, Pd2(dba)3—Tris(dibenzylideneacetone)dipalladium, Pd(dppf)Cl2—(1,1′-Bis(diphenylphosphino)ferrocene)palladium(II) dichloride, Pd(dppf)Cl2·DCM—(1,1-Bis(diphenylphosphino)ferrocene)dichloropalladium(II), complex with DCM, Pd(OAc)2—Palladium(II) acetate, PPh3—Triphenylphosphine, Pet ether—Petroleum ether, PFA—Paraformaldehyde, Pic-BH3—2-Picoline-borane complex, PhI(OAc)2—(Diacetoxyiodo)benzene, Raney Ni—Raney nickel, RF—Retention factor, RT—Room temperature, sat—Saturated, SCX—Strong cation exchange, TBAF—Tetra-n-butylammonium fluoride, TBN—tert-Butyl nitrite, TEA—Triethylamine, THF—Tetrahydrofurane, THP—Tetrahydropyranyl, TFA—Trifluoroacetic acid, TFAA—Trifluoroacetic anhydride, TLC—Thin layer chromatography, TMS—Trimethylsilyl, TMSN3—Trimethylsilyl azide, T3P—Propanephosphonic acid anhydride, Xanthphos—4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene, XPhos—2-Dicyclohexylphosphino-2′,4′,6′-triisopropyl biphenyl, XPhos-Pd-G2—Chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1-biphenyl)[2-(2′-amino-1,1′-biphenyl)]palladium(II), ZnEt2—Diethylzinc.


Compounds of the present invention may be prepared according to the general procedure outlined in Scheme 1.




embedded image


In another embodiment, compounds of the present invention may also be synthesized according to the general procedure outlined in Scheme 2.




embedded image


Alternatively, compounds of the present invention may also be synthesized according to the general procedure outlined in Scheme 3.




embedded image


The general schemes depicted above should be considered as non-limiting examples. It will be understood that compounds of the invention may be obtained through other methods which are known to people skilled in the art.









TABLE 1







Structures of example compounds of the disclosure and their


respective Compound Numbers









Cpd.




No.
Structure
Name












1


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3-yl)methyl)acrylamide





2


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3-yl)methyl)propionamide





3


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3- yl)methyl)methanesulfonamide





4


embedded image


N-((1-(4-(trifluoromethoxy)phenyl)- 1H-indazol-3-yl)methyl)acrylamide





5


embedded image


N-((1-(4-(trifluoromethoxy)phenyl)- 1H-indazol-3-yl)methyl)propionamide





6


embedded image


N-((1-(4-(trifluoromethoxy)phenyl)- 1H-indazol-3- yl)methyl)methanesulfonamide





7


embedded image


2-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3-yl)methyl)isothiazolidine 1,1-dioxide





8


embedded image


N-methyl-1-(4- (trifluoromethyl)phenyl)-1H-indazole- 3-carboxamide





9


embedded image


1-methyl-N-((1-(4- (trifluoromethyl)phenyl)-1H-indazol-3- yl)methyl)-1H-pyrazole-4- sulfonamide





10


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3-yl)methyl)pyridine-3- sulfonamide





11


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3- yl)methyl)cyclopropanesulfonamide





12


embedded image


N-(methylsulfonyl)-1-(4- (trifluoromethyl)phenyl)-1H-indazole- 3-carboxamide





13


embedded image


N,N-dimethyl-N′-((1-(4- (trifluoromethyl)phenyl)-1H-indazol-3- yl)methyl)sulfamide





14


embedded image


N-(cyclopropylsulfonyl)-1-(4- (trifluoromethyl)phenyl)-1H-indazole- 3-carboxamide





15


embedded image


N-(2,3-dihydroxypropyl)-1-(4- (trifluoromethyl)phenyl)-1H-indazole- 3-carboxamide





16


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3- yl)methyl)methanesulfonimidamide





17


embedded image


1-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3-yl)methyl)imidazoiidin-2- one





18


embedded image


3-((1-(4-(trifluoromethyl)phenyl)-1H- indazol-3-yl)methyl)oxazolidin-2-one





19


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- pyrazolo[3,4-b]pyridin-3- yl)methyl)acrylamide





20


embedded image


N-(1-(4-(trifluoromethyl)phenyl)-1H- indol-3-yl)acrylamide





21


embedded image


N-(1-(4-(trifluoromethyl)phenyl)-1H- indol-3-yl)acetamide





22


embedded image


N-(1-phenyl-1H-indol-3-yl)acrylamide





23


embedded image


N-(1-(4-isopropylphenyl)-1H-indol-3- yl)acrylamide





24


embedded image


N-(1-(4-chlorophenyl)-1H-indol-3- yl)acrylamide





25


embedded image


N-(1-(3-chlorophenyl)-1H-indol-3- yl)acrylamide





26


embedded image


N-(1-([1,1′-biphenyl]-4-yl)-1H-indol-3- yl)acrylamide





27


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indol-3-yl)methyl)acrylamide





28


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indol-3-yl)methyl)propionamide





29


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indol-2-yl)methyl)acrylamide





30


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- indol-2-yl)methyl)propionamide





31


embedded image


N-((1-(4-(trifluoromethyl)benzyl)-1H- indol-2-yl)methyl)acrylamide





32


embedded image


N-((1-(3-(trifluoromethyl)benzyl)-1H- indol-2-yl)methyl)acrylamide





33


embedded image


N-(2-oxo-1-(4- (trifluoromethyl)phenyl)indolin-3- yl)acrylamide





34


embedded image


N-(2-oxo-1-(4- (trifluoromethyl)phenyl)indolin-3- yl)acetamide





35


embedded image


2-(8-(1-methyl-1H-pyrazol-3-yl)-6- phenylimidazo[1,2-a]pyridin-2- yl)acetic acid





36


embedded image


2-(6-phenyl-8-(1-(pyridin-3-ylmethyl)- 1H-pyrazol-4-yl)imidazo[1,2- a]pyridin-2-yl)acetic acid





37


embedded image


N-methyl-2-(8-(1-methyl-1H-pyrazol- 3-yl)-6-phenylimidazo[1,2-a]pyridin-2- yl)acetamide





38


embedded image


2-(8-(1-methyl-1H-pyrazol-3-yl)-6- (phenylamino)imidazo[1,2-a]pyridin- 2-yl)acetic acid





39


embedded image


N-methyl-2-(8-(1-methyl-1H-pyrazol- 3-yl)-6-(phenylamino)imidazo[1,2- a]pyridin-2-yl)acetamide





40


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- pyrazolo[4,3-b]pyridin-3- yl)methyl)acrylamide





41


embedded image


N-((1-(4-(trifluoromethyl)phenyl)-1H- pyrazolo[4,3-c]pyridin-3- yl)methyl)acrylamide





42


embedded image


N-[[5-(cyanomethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





43


embedded image


N-[[5-(cyanomethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





44


embedded image


N-[[5-[[rac-tetrahydrofuran-3- yl]methoxy]-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





45


embedded image


N-[[5-phenoxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





46


embedded image


N-[[5-[[rac-tetrahydrofuran-3- yl]methoxy]-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





47


embedded image


N-[[5-[[(3S)-tetrahydrofuran-3- yl]methoxy]-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





48


embedded image


N-[[5-[[(3R)-tetrahydrofuran-3- yl]methoxy]-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





49


embedded image


N-[[5-phenoxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





50


embedded image


N-[[6-(cyclopropoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





51


embedded image


N-[[5-(3-methyl-2,6-dioxo-pyrimidin- 4-yl)oxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





52


embedded image


N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





53


embedded image


N-[[5-(3-methyl-2,6-dioxo-pyrimidin- 4-yl)oxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]propanamide





54


embedded image


N-[[5-benzyloxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





55


embedded image


N-[[5-benzyloxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]propanamide





56


embedded image


N-[[5-benzyloxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





57


embedded image


1-cyano-N-[[1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





58


embedded image


N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





59


embedded image


N-[[1-[4- (trifluoromethyl)phenyl]pyrazolo[3,4- c]pyridin-3-yl]methyl]prop-2-enamide





60


embedded image


N-[[5-(3-methyl-2,6-dioxo-pyrimidin- 4-yl)oxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





61


embedded image


N-[[6-(cyclopropoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





62


embedded image


N-[[6-(cyclopropoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]propanamide





63


embedded image


N-[[5-prop-2-ynoxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





64


embedded image


N-[[5-prop-2-ynoxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]propanamide





65


embedded image


N-[[5-prop-2-ynoxy-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





66


embedded image


N-[[5-(cyclopropylmethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





67


embedded image


N-[[5-(cyclopropylmethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]propanamide





68


embedded image


N-[[5-(cyclopropylmethoxy)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





69


embedded image


2-[1-[4- (trifluoromethyl)phenyl]indazol-4- yl]oxyacetic acid





70


embedded image


2-[[[1-[4- (trifluoromethyl)phenyl]indazol-3- yl]amino]methyl]prop-2-enoic acid





71


embedded image


N-[[5-(trifluoromethyl)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





72


embedded image


N-[[5-(trifluoromethyl)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]acetamide





73


embedded image


N-[[5-(trifluoromethyl)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





74


embedded image


N-[[6-(trifluoromethyl)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]prop-2-enamide





75


embedded image


N-[[6-(trifluoromethyl)-1-[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]acetamide





76


embedded image


N-[[6-(trifluoromethyl)-1 -[4- (trifluoromethyl)phenyl]indazol-3- yl]methyl]methanesulfonamide





77


embedded image


N-[[1-(4,4-difluorocyclohexyl)indazol- 3-yl]methyl]prop-2-enamide





78


embedded image


N-[[4-[(4-fluorophenyl)methoxy]-1- methyl-indol-6-yl]methyl]prop-2- enamide





79


embedded image


N-[[4-[(4-fluorophenyl)methoxy]-1- methyl-indol-6-yl]methyl]acetamide





80


embedded image


N-[[4-(4-chlorophenoxy)-1-methyl- indol-6-yl]methyl]acetamide





81


embedded image


N-[[4-(4-chlorophenoxy)-1-methyl- indol-6-yl]methyl]prop-2-enamide





82


embedded image


N-[[4-[(4-fluorophenyl)methoxy]-1-[(2- oxo-1H-pyridin-4-yl)methyl]indol-6- yl]methyl]prop-2-enamide





83


embedded image


N-[[4-[(4-fluorophenyl)methoxy]-1-[(2- oxo-1H-pyridin-4-yl)methyl]indol-6- yl]methyl]acetamide





84


embedded image


N-[[4-(4-chlorophenoxy)-1-[(2-oxo- 1H-pyridin-4-yl)methyl]indol-6- yl]methyl]prop-2-enamide





85


embedded image


N-[[4-(4-chlorophenoxy)-1-[(2-oxo- 1H-pyridin-4-yl)methyl]indol-6- yl]methyl]acetamide





86


embedded image


2-[[[4-[(4-fluorophenyl)methoxy]-1- methyl-indol-6- yl]methylamino]methyl]prop-2-enoic acid





87


embedded image


2-[[[4-(4-chlorophenoxy)-1-methyl- indol-6-yl]methylamino]methyl]prop- 2-enoic acid









These examples are provided for the purpose of illustrating the present disclosure and by no means should be interpreted to limit the scope of the present disclosure.


Part A: Experimental Chemistry Procedures

All starting materials which are not explicitly described were either commercially available (the details of suppliers such as for example Acros, Avocado, Aldrich, Fluka, FluoroChem, MatrixScientific, Maybridge, Merck, Sigma, etc. can be found in the SciFinder® Database for example) or the synthesis thereof has already been described precisely in the specialist literature (experimental guidelines can be found in the Reaxys® Database or the SciFinder® Database respectively, for example) or can be prepared using the conventional methods known to the person skilled in the art.


The reactions were, if necessary, carried out under an inert atmosphere (mostly argon and N2). The number of equivalents of reagents and the amounts of solvents employed as well as the reaction temperatures and times can vary slightly between different reactions carried out by analogous methods. The work-up and purification methods were adapted according to the characteristic properties of each compound and can vary slightly for analogous methods. The yields of the compounds prepared are not optimized.


The indication “equivalents” (“eq.” or “eq” or “equiv.”) means molar equivalents, “RT” or “rt” means room temperature T (23±7° C.), “M” are indications of concentration in mol/l, “sol.” means solution, “conc.” means concentrated. The mixing ratios of solvents are usually stated in the volume/volume ratio.


To perform reactions under microwave radiation a CEM Microwave (Discover SP) was employed (Heating rate: 2-6° C./sec; Temperature: 30-300° C. volume-independent infrared (IR) and 80-300° C. Fiber optic (FO) temperature measurement; Pressure: 0-435 psi, ActiVent™ technology; Power: 0-300 W; Magnetron frequency: 2450 MHz; Reaction agitation: electromagnetic stirring; Air Cooling: 25 psi (20 L/min flow); System control: Synergy™ software).


Key analytical characterization was carried out by means of 1H-NMR spectroscopy and/or mass spectrometry (MS, m/z for [M+H]+ and/or [M−H]) for all the exemplary compounds and selected intermediate products. In certain cases, where e.g. regioisomers and/or diatereomers could be/were formed during the reaction, additional analytics, such as, e.g. 13C NMR and NOE (nuclear overhauser effect) NMR experiments were in some cases performed.


Analytical instruments employed were e.g. for NMR analysis a BRUKER AVANCE 400 MHz (Software Topspin) or a VARIAN MR 400 MHz (VNMRJ Sofware) machine was employed. For LC/MS analysis e.g. an Acquity UPLC H-Class, Mass: Acquity SQD2 Detector (ESI), an Acquity UPLC, Mass: Quatro premier XE Detector (ESI), an Acquity UPLC, Mass: Waters Xevo TQ-S Detector (ESI/ESCI), or an Alliance Waters 2695, Mass: Quattromicro™ (ESCI) multimode ionization was employed. Analytical HPLCs were measured e.g. on Alliance Waters 2695). Analytical SFC were performed e.g. on a PIC solution (Software: SFC PIC Lab Online), a WATERS-X5 (Software MASSLYNX), or a WATERS-UPC2 (Empower).


Preparative HPLC were performed e.g. on a Waters 2545 (Software Empower), a Gilson (Software Trilution), or a Shimadzu (Software LC Solution). Preparative SFC were performed e.g. on a Waters Thar SFC-80 (Software Chromscope), Waters Thar SFC-150 (Software Chromscope), Waters Thar SFC-200 (Software Chromscope), or a PIC SFC-175 (Software SFC PIC Lab Online).


Structures of example compounds that contain stereocentres are drawn and named with absolute stereochemistry, if known. In case of unknown absolute stereochemistry the compounds can be either racemic, a mixture of diatereomers, a pure diastereomer of unknown stereochemistry, or a pure enantiomer of unknown stereochemistry. Dia 1 and Dia 2 means that diastereiosomers were separated but the stereochemistry is unknown. En 1 and En 2 means that both enantiomers were separated but the absolute configuration is unknown. No suffix given after the compound code means that a compound containing stereocentres was obtained as a racemic mixture or a mixture of diatereomers, respectively, unless the chemical name of the compound specifies the exact stereochemistry.


The LC/MS analysis mentioned in the experimental part were performed on a Alliance Waters 2695 HPLC (equipped with a PDA detector) connected to a mass spectrometer Waters Quattromicro (ESCI, multimode ionization). The separations were performed with a Acquity BEH C18 (1.7 μm, 2.1×50 mm) column, a Acquity BEH C18 (1.7 μm, 2.1×100 mm) column, or a X-Bridge C18 (3.5 μm, 4.6×75 mm) column thermostated to 30-35° C. and the PDA acquisition wavelength was set in the range of 210-400 nm (Acuisition Software: MassLynx). Elutions were carried out with the methods described in the following table. For Methods L1, L6, and L7 Solvent A: FA LC-MS grade 0.1% in milliQ water. Solvent B: FA LC-MS grade 0.1% in ACN LC-MS grade. For Methods L2 and L3 Solvent A: FA LC-MS grade 0.05% in milliQ water. Solvent B: FA LC-MS grade 0.05% in ACN LC-MS grade. For Methods L4 and L8 Solvent A: TFA LC-MS grade 0.05% in milliQ water. Solvent B: TFA LC-MS grade 0.05% in ACN LC-MS grade. For Method L5, Solvent A: 5 mM (NH4)HCO3 in milliQ water. Solvent B: ACN LC-MS grade. For Method L9, Solvent A: TFA LC-MS grade 0.1% in milliQ water. Solvent B: ACN LC-MS grade.









TABLE 2







HPLC methods











HPLC
Time
Solvents
Flow














Method
System
(min)
A (%)
B (%)
(mL/min)
Column
















L1
Alliance Waters
0
97
3
0.6
Acquity BEH



2695 HPLC
0.4
97
3
0.6
C18 (1.7 μm,




3.2
2
98
0.6
2.1 × 50 mm)




3.8
2
98
0.6
(0.1% FA in




4.2
97
3
0.6
solvents A and




4.5
97
3
0.6
B)


L2
Alliance Waters
0
97
3
0.6
Acquity BEH



2695 HPLC
0.4
97
3
0.6
C18 (1.7 μm,




3.2
2
98
0.6
2.1 × 50 mm)




3.8
2
98
0.6
(0.05% FA in




4.2
97
3
0.6
solvents A and




4.5
97
3
0.6
B)


L3
Alliance Waters
0
97
3
0.6
Acquity BEH



2695 HPLC
0.4
97
3
0.6
C18 (1.7 μm,




7.5
2
98
0.6
2.1 × 50 mm)




9.5
2
98
0.6
(0.05% FA in




9.6
97
3
0.6
solvents A and




10
97
3
0.6
B)


L4
Alliance Waters
0
97
3
0.6
Acquity BEH



2695 HPLC
0.4
97
3
0.6
C18 (1.7 μm,




2.5
2
98
0.6
2.1 × 100 mm)




3.4
2
98
0.6
(0.05% TFA in




3.5
97
3
0.6
solvents A and




4
97
3
0.6
B)


L5
Alliance Waters
0
95
5
1.3
X-Bridge C18



2695 HPLC
0.5
95
5
1.3
(3.5 μm,




1.0
85
15
1.3
4.6 × 75 mm)




4.0
2
98
1.3
(5 mM




7.0
2
98
1.3
(NH4)HCO3 in




7.5
95
5
1.3
solvent A)




8.0
95
5
1.3


L6
Alliance Waters
0
97
3
0.6
Acquity BEH



2695 HPLC
0.4
97
3
0.6
C18 (1.7 μm,




2.5
2
98
0.6
2.1 × 50 mm)




3.4
2
98
0.6
(0.05% FA in




3.5
97
3
0.6
solvents A and




4.0
97
3
0.6
B)


L7
Alliance Waters
0
97
3
0.55
Acquity BEH



2695 HPLC
8.5
0
100
0.55
C18 (1.7 μm,




9.0
0
100
0.55
2.1 × 100 mm)




9.5
97
3
0.55
(0.1% FA in




10.0
97
3
0.55
solvents A and B)


L8
Alliance Waters
0
97
3
0.45
Acquity BEH



2695 HPLC
0.4
97
3
0.45
C18 (1.7 μm,




3.5
2
98
0.45
2.1 × 100 mm)




4.5
2
98
0.45
(0.05% FA in




5.0
3
97
0.45
solvents A and




5.5
3
97
0.45
B)


L9
Alliance Waters
0
97
3
0.55
Acquity BEH



2695 HPLC
8.5
0
100
0.55
C18 (1.7 μm,




9.5
0
100
0.55
2.1 × 100 mm)




10
97
3
0.55
(0.1% TFA in








solvent A)









The UPLC analysis mentioned in the experimental part were performed on a Acquity UPLC H-Class (equipped with a PDA detector) connected to a mass spectrometer Acquity TQ Detector (ESI/ESCI). The separations were performed with a Acquity UPLC BEH C18 (1.7 μm, 2.1×50 mm) or Acquity UPLC HSS C18 (1.8 μm, 2.1×50 mm) column thermostated to 40° C. and the PDA acquisition wavelength was set in the range of 190-420 nm. Elutions were carried out with the methods described in the following table. For Methods U1 and U2 Solvent A: 10 mM (NH4)OAc in milliQ water containing 5% ACN (UPLC grade, Biosolve). Solvent B: ACN (UPLC grade, Biosolve). For Method U3 Solvent A: 0.1% FA (UPLC grade, Biosolve) in milliQ water. Solvent B: ACN (UPLC grade, Biosolve).









TABLE 3







UPLC methods











UPLC
Time
Solvents
Flow














Method
System
(min)
A (%)
B (%)
(mL/min)
Column
















U1
Acquity UPLC
0
84
16
0.5
Acquity UPLC



H-Class
3.4
42
58
0.5
BEH C18




4.0
10
90
0.5




5.0
10
90
0.5


U2
Acquity UPLC
0
52
48
0.5
Acquity UPLC



H-Class
3.5
10
90
0.5
BEH C18




5.0
10
90
0.5


U3
Acquity UPLC
0
80
20
0.5
Acquity UPLC



H-Class
3.4
40
40
0.5
HSS C18




4.0
10
90
0.5




5.0
10
90
0.5









Preparative HPLC purifications mentioned in this experimental part have been carried out on a Waters 2545 (Empower software, 2996 PDA detector, 2707 autosampler), a Gilson (Software Trilution, 171 DAD detector, GX-271 autosampler), or a Shimadzu (Software LC Solution, CMB-20A detector, SIL-10AP autosampler). The separations were performed with a Luna C18 (5 μm, 21.2×250 mm; or 10 μm, 21.2×250 mm) column, a X-Bridge C18 (5 μm, 29×250 mm; 5 μm, 19×150 mm; or 10 μm, 19×150 mm) column, a X-Select C18 (5 μm, 19×150 mm; 5 μm, 19×250 mm; 5 μm, 25×150 mm; 10 μm, 25×150 mm; or 10 μm, 25×250 mm) column, a X-Select CSH Phenyl-Hexyl (5 μm, 19×150 mm; or 5 μm, 19×250 mm) column, a Synergi Polar-RP (5 μm, 21.2×250 mm) column, a Kromasil (5 μm, 19×150 mm) column, a YMC-Triart C18 (10 μm, 19×250 mm) column, or a SUNFIRE C18 (5 μm, 19×250 mm) column. Elutions were carried out with columns and solvents described in the following table. Gradients systems for each individual compound were employed using the solvents mentioned in the table. Detection wavelengths were fixed at 210 and 254 nm.









TABLE 4







Preparative HPLC methods








HPLC



Method
Column and conditions





H1
Luna C18 (5 μm, 21.2 × 250 mm) or Luna C18 (10 μm, 21.2 × 250 mm); Solvent A:



10 mM (NH4)HCO3 in water; Solvent B: ACN; Flow: 15 mL/min.


H2
X-Bridge C18 (5 μm, 19 × 150 mm), X-Bridge C18 (5 μm, 19 × 250 mm), or X-Bridge



C18 (10 μm, 19 × 150 mm); Solvent A: 10 mM (NH4)HCO3 in water; Solvent B: ACN;



Flow: 16 mL/min.


H3
X-Select C18 (5 μm, 19 × 150 mm), X-Select C18 (5 μm, 19 × 250 mm), X-Select C18



(10 μm, 25 × 150 mm), or X-Select C18 (10 μm, 25 × 250 mm); Solvent A: 10 mM



(NH4)HCO3 in water; Solvent B: ACN; Flow: 15 mL/min.


H4
X-Select C18 (5 μm, 19 × 250 mm); Solvent A: 10 mM (NH4)HCO3 in water; Solvent



B: ACN/MeOH (1:1); Flow: 13 mL/min.


H5
X-Select CSH Phenyl-Hexyl (5 μm, 19 × 150 mm), X-Select CSH Phenyl-Hexyl (5 μm,



19 × 250 mm) or X-Select CSH Phenyl-Hexyl (10 μm, 21.2 × 250 mm); Solvent A:



10 mM (NH4)HCO3 in water; Solvent B: ACN; Flow: 13 mL/min.


H6
X-Select C18 (5 μm, 20 × 250 mm) or X-Select C18 (10 μm, 25 × 150 mm); Solvent A:



10 mM NH4OAc in water; Solvent B: ACN; Flow: 20 mL/min.


H7
X-Select CSH Phenyl-Hexyl (5 μm, 19 × 250 mm); Solvent A: 10 mM NH4OAc in



water; Solvent B: ACN; Flow: 16 mL/min.


H8
X-Select CSH Phenyl-Hexyl (5 μm, 19 × 250 mm); Solvent A: 0.1% FA in water;



Solvent B: ACN; Flow: 14 mL/min.


H9
X-Select C18 (5 μm, 25 × 150 mm), X-Select C18 (10 μm, 25 × 150 mm) or X-Select



C18 (5 μm, 19 × 250 mm); Solvent A: 0.1% FA in water; Solvent B: ACN; Flow: 18



mL/min.


H10
X-Bridge C18 (5 μm, 19 × 150 mm); Solvent A: 0.1% AcOH in water; Solvent B: ACN;



Flow: 14 mL/min.


H11
Synergi Polar-RP (5 μm, 21.2 × 250 mm); Solvent A: 0.1% FA in water; Solvent B:



ACN; Flow: 17 mL/min.


H12
Kromasil (5 μm, 19 × 150 mm); Solvent A: 10 mM (NH4)HCO3 in water; Solvent B:



ACN; Flow: 15 mL/min.


H13
Luna C18 (5 μm, 21.2 × 250 mm); Solvent A: 0.1% FA in water; Solvent B: ACN; Flow:



15 mL/min.


H14
YMC-Triart C18 (10 μm, 19 × 250 mm); Solvent A: 10 mM (NH4)HCO3 in water;



Solvent B: ACN; Flow: 20 mL/min.


H15
YMC-Triart C18 (10 μm, 19 × 250 mm); Solvent A: 10 mM (NH4)HCO3 in water;



Solvent B: ACN/MeOH (1:1); Flow: 22 mL/min.


H16
X-Select CSH Phenyl-Hexyl (5 μm, 25 × 150 mm); Solvent A: 0.1% TFA in water;



Solvent B: ACN; Flow: 15 mL/min.


H17
SUNFIRE C18 (5 μm, 19X250 mm); Solvent A: 0.1% FA in water; Solvent B: ACN;



Flow: 17 mL/min.









Example 1: Synthesis of N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acrylamide (Cpd. No. 001)



embedded image


Step 1: To a mixture of 3-(aminomethyl)-1H-indazole (100 mg, 0.65 mmol) in MeOH (4 mL) was added aq 1M NaHCO3 (1.3 mL, 1.29 mmol) followed by Boc2O (145 mg, 0.65 mmol). The reaction mixture was stirred at RT for 30 min. Progress of the reaction was followed by LC/MS. The mixture was concentrated, diluted with H2O and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 12 g column (silica) and a gradient of 10-80% EtOAc in n-heptane as an eluent to afford tert-butyl (1H-indazol-3-ylmethyl)carbamate (75 mg, 47%). (LC/MS; m/z 246 [M−H]).


Step 2: A reaction vial was charged with tert-butyl (1H-indazol-3-ylmethyl)carbamate (60 mg, 0.24 mmol), 4-iodobenzotrifluoride (44 μL, 0.30 mmol), K3PO4 (132 mg, 0.60 mmol), 2-picolinic acid (4.0 mg, 0.032 mmol) and CuI (6.0 mg, 0.032 mmol). The vial was purged with argon gas and DMSO (1.5 mL) was added. The reaction mixture was stirred at 80° C. for 16 h (sealed tube). Progress of the reaction was followed by LC/MS. The mixture was diluted with EtOAc, filtered through a celite pad and washed with EtOAc. The filtrate was washed with sat NH4Cl, a NH4Cl/NaOH (pH 8) solution and brine. The solution was dried over MgSO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 12 g column (silica) and a gradient of 5-40% EtOAc in n-heptane as an eluent to afford tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate as a white solid (60 mg, 63%). (LC/MS; m/z 336 [M+H−tBu]+).


Step 3: A solution of tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (60 mg, 0.15 mmol) in 1,4-dioxane (0.6 mL) was treated with HCl (4M in 1,4-dioxane; 0.6 mL, 2.40 mmol). The reaction mixture was stirred at RT for 16 h. Progress of the reaction was followed by LC/MS. The mixture was concentrated under reduced pressure to afford (1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine hydrochloride as a white solid (54 mg, 107%). The crude product was used without further purification in the next step.


Step 4: To a suspension of (1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine hydrochloride (30 mg, 0.092 mmol) in DCM (0.6 mL) was added TEA (51 μL, 0.37 mmol) followed by acryloyl chloride (10 μL, 0.12 mmol). The reaction mixture was stirred at RT for 15 min. The mixture was concentrated under reduced pressure and the residue was purified by normal phase flash column chromatography using a 12 g column (silica) and a gradient of 20-80% EtOAc in n-heptane as an eluent to afford N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acrylamide (Cpd. No. 001) as a white solid (19 mg, 60%). (LC/MS; m/z 346 [M+H]+).


The following compounds were prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 001: Cpd. No. 002 (employing propionyl chloride at step 4), Cpd. No. 003 (employing mesyl chloride at step 4), Cpd. No. 004 (employing 4-trifluoromethoxyphenyl iodide at step 2), Cpd. No. 005 (employing 4-trifluoromethoxyphenyl iodide at step 2 and propionyl chloride at step 4), Cpd. No. 006 (employing 4-trifluoromethoxyphenyl iodide at step 2 and mesyl chloride at step 4), Cpd. No. 057 (employing cyanomethylsulfonyl chloride at step 4).


Example 2: Synthesis of N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acrylamide (Cpd. No. 020)



embedded image


Step 1: A solution of methyl 1H-indole-3-carboxylate (1.0 g, 5.7 mmol), 1-iodo-4-(trifluoromethyl)benzene (3.87 g, 17.1 mmol), Cs2CO3 (3.71 g, 11.4 mmol) and Cu2O (815 mg, 5.7 mmol) in DMA (7 mL) was stirred at 200° C. for 1 h (sealed tube; microwave irradiation). Progress of the reaction was monitored by TLC. TLC mobile phase: 10% EtOAc in pet ether, RF: 0.5, TLC detection: UV. The reaction mixture was cooled to RT, diluted with EtOAc (80 mL) and washed with H2O (50 mL). The organic layer was washed with brine (40 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product (1.2 g) was purified by normal phase flash column chromatography using silica gel (100-200 mesh) and a gradient of 0-5% of EtOAc in pet ether as an eluent to afford methyl 1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carboxylate as an off-white solid (625 mg, 33%, LC/MS 96%). (LC/MS; m/z 320 [M+H]+).


Step 2: A solution of methyl 1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carboxylate (2.5 g, 7.8 mmol) and LiOH·H2O (2.62 g, 62.4 mmol) in MeOH (25 mL), THF (25 mL) and H2O (12.5 mL) was stirred at 80° C. for 4 h. Progress of the reaction was monitored by TLC. TLC mobile phase: 30% EtOAc in pet ether, RF: 0.5, TLC detection: UV. The reaction mixture was concentrated under reduced pressure and acidified with aq 2M HCl (pH˜2) at 0° C. The precipitated solids were filtered and dried under vacuum to afford 1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carboxylic acid as an off-white solid (1.8 g, 73%, LC/MS 96%). (LC/MS; m/z 306 [M+H]+).


Step 3: A solution of 1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carboxylic acid (1.3 g, 4.3 mmol) in THF (20 mL) was treated with DPPA (2.95 g, 10.75 mmol) and TEA (1.30 g, 12.9 mmol) and stirred at RT for 2 h. The reaction mixture was concentrated under reduced pressure, the residue was dissolved in tBuOH (25 mL) and the solution was stirred at 110° C. for 16 h. Progress of the reaction was monitored by TLC. TLC mobile phase: 50% EtOAc in pet ether, RF: 0.76, TLC detection: UV. The reaction mixture was diluted with EtOAc (100 mL) and washed with H2O (60 mL). The organic layer was washed with brine (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product (1.3 g) was purified by normal phase flash column chromatography using silica gel (100-200 mesh) and a gradient of 0-7% of EtOAc in pet ether to afford tert-butyl (1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)carbamate as an off-white solid (600 mg, 37%, LC/MS 97%). (LC/MS; m/z 377 [M+H]+).


Step 4: A solution of tert-butyl (1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)carbamate (300 mg, 0.79 mmol) in DCM (7 mL) was treated with HCl (4M in 1,4-dioxane) (2 mL) and stirred at RT for 6 h. Progress of the reaction was monitored by TLC. TLC mobile phase: 50% EtOAc in pet ether, RF: 0.1, TLC detection: UV. The reaction mixture was concentrated under reduced pressure to afford 1-(4-(trifluoromethyl)phenyl)-1H-indol-3-amine hydrochloride as a pale yellow solid (150 mg, 24%, LC/MS 34%). (LC/MS; m/z 277 [M+H]+). The product was used as such without further purification.


Step 5: A solution of 1-(4-(trifluoromethyl)phenyl)-1H-indol-3-amine hydrochloride (100 mg, 0.32 mmol, LC/MS 34%) and NaHCO3 (54 mg, 0.63 mmol) in 1,4-dioxane (3 mL) and water (1 mL) was treated with a solution of acryloyl chloride (35 mg, 0.38 mmol) in 1,4-dioxane (2 mL) at 0° C. and stirred for 10 min. Progress of the reaction was monitored by TLC. TLC mobile phase: 30% of EtOAc in pet ether, RF: 0.61, TLC detection: UV. The reaction mixture was diluted with DCM (10 mL) and washed with H2O (7 mL). The organic layer was washed with brine (7 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product (110 mg, LC/MS 73%) was purified by preparative HPLC method H9. The collected fractions were lyophilized to afford N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acrylamide (Cpd. No. 022) as an off-white solid (21 mg, 58%, LC/MS 99%). (LC/MS; m/z 331 [M+H]+).


The following compounds were prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 020: Cpd. No. 021 (employing Ac2O and DIPEA in DCM at step 5), Cpd. No. 022, Cpd. No. 023, Cpd. No. 024, Cpd. No. 025, Cpd. No. 026.


Example 3: Synthesis of N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)acrylamide (Cpd. No. 027)



embedded image


Step 1: A solution of 1H-indole-3-carbaldehyde (1.0 g, 6.9 mmol), K2CO3 (1.99 g, 14.49 mmol) and 1-iodo-4-(trifluoromethyl)benzene (3.75 g, 13.8 mmol) in 1,4-dioxane (10 ml) was degassed with argon for 10 min. The mixture was treated with CuI (262 mg, 1.38 mmol) and (±)-trans-1,2-diaminocyclohexane (157 mg, 1.38 mmol) and stirred at 120° C. for 16 h (sealed tube). Progress of the reaction was monitored by TLC. TLC mobile phase: 10% EtOAc in pet ether, RF: 0.5, TLC detection: UV. The reaction mixture was diluted with EtOAc (100 ml) and washed with brine (3×70 ml). The combined organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product (1.2 g, LC/MS 78%) was purified by normal phase flash column chromatography using silica gel (100-200 mesh) and a gradient of 0-5% EtOAc in pet ether to afford 1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carbaldehyde as an off-white solid (980 mg, 49%, LC/MS 99%). (LC/MS; m/z 290 [M+H]+).


Step 2: A solution of 1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carbaldehyde (250 mg, 0.9 mmol), NH2OH·HCl (93 mg, 1.35 mmol) and Na2CO3 (71 mg, 0.675 mmol) in EtOH (10 mL) and H2O (1 mL) was stirred at 100° C. for 2 h. Progress of the reaction was monitored by TLC. TLC mobile phase: 10% EtOAc in pet ether, RF: 0.35, TLC detection: UV. The reaction mixture was diluted with EtOAc (100 mL) and washed with brine (2×50 mL). The organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford (E/Z)-1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carbaldehyde oxime as an off-white solid (230 mg, 86%, LC/MS 36% & 62%). (LC/MS; m/z 305 [M+H]+).


Step 3: A solution of (E/Z)-1-(4-(trifluoromethyl)phenyl)-1H-indole-3-carbaldehyde oxime (230 mg, 0.75 mmol) and NiCl2.6H2O (182 mg, 0.77 mmol) in MeOH (10 ml) was treated with NaBH4 (319 mg, 8.4 mmol) at 0° C. The reaction mixture was stirred at 0° C. for 2 h. Progress of the reaction mixture was monitored by TLC. TLC mobile phase: 10% MeOH in DCM, RF: 0.6, TLC detection: UV. The reaction mixture was diluted with EtOAc (50 mL), stirred for 10 min and filtered through a celite pad. The filtrate was washed with H2O (20 mL) and brine (2×25 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford (1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methanamine as a brown gum (180 mg, 48%, LC/MS 57%). (LC/MS; m/z 291 [M+H]+).


Step 4: A solution of (1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methanamine (90 mg, 0.31 mmol, LC/MS 57%) and NaHCO3 (78 mg, 0.92 mmol) in 1,4-dioxane (5 mL) and H2O (1 mL) was cooled to 0° C. and treated with acryloyl chloride (28 mg, 0.31 mmol). The reaction mixture was stirred at 0° C. for 1 h. Progress of the reaction mixture was monitored by TLC. TLC mobile phase: 10% EtOAc in pet ether, RF: 0.5, TLC detection: UV. The reaction mixture was diluted with DCM (30 mL) and washed with H2O (15 ml) and brine (2×20 mL). The organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product (130 mg, LC/MS 38%) was purified by preparative HPLC method H2. The collected fractions were lyophilized to afford N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)acrylamide (Cpd. No. 027) as a white solid (21 mg, 34%, LC/MS 99%). (LC/MS; m/z 345 [M+H]+).


The compound Cpd. No. 028 (employing propionyl chloride and TEA in DCM at step 4) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 027.


Example 4: Synthesis of N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)acrylamide (Cpd. No. 029)



embedded image


Step 1: A solution of (1H-indol-2-yl)methanamine hydrochloride (2.0 g, 11 mmol), TEA (2.22 g, 22 mmol) and DMAP (67 mg, 0.55 mmol) in DCM (40 mL) was treated with Boc2O (2.39 g, 11 mmol). The reaction mixture was stirred at RT for 3 h. Progress of the reaction mixture was monitored by TLC. TLC mobile phase: 60% EtOAc in pet ether, RF: 0.7, TLC detection: UV. The reaction mixture was diluted DCM (30 mL) and washed with H2O (20 mL). The organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 40 g column (silica) and a gradient of 0-10% EtOAc in pet ether as an eluent to afford tert-butyl ((1H-indol-2-yl)methyl)carbamate (Int-1) as an off-white solid (2.0 g, 74%, LC/MS 99%). (LC/MS; m/z 247 [M+H]+).


Step 2: A solution of Int-1 (1.0 g, 4.06 mmol), 1-iodo-4-(trifluoromethyl)benzene (2.23 g, 8.13 mmol), K3PO4 (1.73 g, 8.13 mmol), CuI (389 mg, 2.03 mmol) and trans-1,2-diaminocyclohexane (93 mg, 0.82 mmol) in toluene (10 mL) was stirred at 120° C. for 16 h under a nitrogen atmosphere (sealed tube). Progress of the reaction mixture was monitored by TLC. TLC mobile phase: 10% EtOAc in pet ether, RF: 0.33, TLC detection: UV. The reaction mixture was concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 24 g column (silica) and a gradient of 10% EtOAc in pet ether as an eluent to afford tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)carbamate as an off-white solid (400 mg, 24%, LC/MS 95%). (LC/MS; m/z 391 [M+H]+).


Steps 3-4: These steps were executed in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to step 4 and 5 towards Cpd. No. 020. Starting material (400 mg, 1.02 mmol, LC/MS 95%) yielded crude product (385 mg, LC/MS 68%) which was purified preparative HPLC method H5. The obtained fractions were lyophilized to afford N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)acrylamide (Cpd. No. 029) as an off-white solid (98 mg, 29%, LC/MS 99%). (LC/MS; m/z 345 [M+H]+).


The compound Cpd. No. 030 (employing propionyl chloride and TEA in DCM at step 4) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 029.


Example 5: Synthesis of N-((1-(4-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide (Cpd. No. 031)



embedded image


Step 1: A solution of Int-1 (300 mg, 1.22 mmol, LC/MS 91%), K2CO3 (336 mg, 2.44 mmol) and 1-(bromomethyl)-4-(trifluoromethyl)benzene (232 mg, 0.975 mmol) in ACN (6 mL) was stirred at 80° C. for 16 h. Progress of the reaction was monitored by TLC. TLC mobile phase: 20% EtOAc in pet ether, RF: 0.5, TLC detection: UV. The reaction mixture was diluted with H2O (20 mL) and extracted with EtOAc (2×20 mL). The combined organic layer was washed with H2O (10 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product (450 mg, LC/MS 35%) was purified by normal phase flash column chromatography using a 12 g column (silica) and a gradient of 0-20% EtOAc in pet ether as an eluent to afford tert-butyl ((1-(4-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)carbamate (120 mg, 16%, LC/MS 60%). (LC/MS; m/z 405 [M+H]+).


Steps 2-3: These steps were executed in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to step 4 and 5 towards Cpd. No. 020. Starting material (120 mg, 0.30 mmol, LC/MS 60%) yielded crude product (120 mg, LC/MS 19%) which was purified preparative HPLC method H2. The obtained fractions were lyophilized to afford N-((1-(4-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide (Cpd. No. 031) as a white solid (7 mg, 11%, LC/MS 99%). (LC/MS; m/z 359 [M+H]+).


The compound Cpd. No. 032 was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 031.


Example 6: Synthesis of N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acrylamide (Cpd. No. 033)



embedded image


Step 1: A solution of 1-(2-aminophenyl)ethan-1-one (4.8 g, 35.5 mmol), Cu(OAc)2 (7.74 g, 42.7 mmol), DIPEA (18.9 mL, 106.6 mmol) and (4-(trifluoromethyl)phenyl)boronic acid (10.1 g, 53.3 mmol) in DCM (150 mL) was stirred under an oxygen atmosphere (balloon pressure) for 16 h at RT. Progress of the reaction was monitored by TLC. TLC mobile phase: 10% EtOAc in pet ether, RF: 0.49, TLC detection: UV. The reaction mixture was diluted with H2O (50 mL) and extracted with DCM (100 mL). The organic layer was washed with H2O (2×30 mL) and brine (30 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product (6.3 g, LC/MS 46%) was purified by normal phase flash column chromatography using a 40 g column (silica) and a gradient of 0-8% EtOAc in pet ether as an eluent to afford 1-(2-((4-(trifluoromethyl)phenyl)amino)phenyl)ethan-1-one as a yellow solid (2.8 g, 26%, LC/MS 92%). (LC/MS; m/z 280 [M+H]+).


Step 2: A solution of 1-(2-((4-(trifluoromethyl)phenyl)amino)phenyl)ethan-1-one (2.3 g, 8.24 mmol) and SeO2 (4.57 g, 41.2 mmol) in 1,4-dioxane (25 mL) was stirred at 80° C. for 16 h under an oxygen atmosphere (balloon pressure). Progress of the reaction was monitored by TLC. TLC mobile phase: 20% EtOAc in pet ether, RF: 0.15, TLC detection: UV. The reaction mixture was diluted with EtOAc (100 mL), filtered through a celite pad and the filtrate was concentrated under reduced pressure. The crude product (3.2 g) was purified by normal phase flash column chromatography using a 40 g column and a gradient of 0-12% EtOAc in pet ether as an eluent to afford 1-(4-(trifluoromethyl)phenyl)indoline-2,3-dione as a red solid (1.0 g, 33%, LC/MS 73%). (LC/MS; m/z 292 [M+H]+).


Step 3: A solution of 1-(4-(trifluoromethyl)phenyl)indoline-2,3-dione (1.0 g, 3.43 mmol), NH2OH·HCl (358 mg, 5.15 mmol) and NaOAc (422 mg, 5.15 mmol) in EtOH (30 mL) was stirred at 90° C. for 4 h. Progress of the reaction was monitored by TLC. TLC mobile phase: 30% EtOAc in pet ether, RF: 0.15, TLC detection: UV. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in EtOAc (100 mL) and H2O (50 mL). The organic layer was dried over Na2SO4 and concentrated under reduced pressure to afford 3-(hydroxyimino)-1-(4-(trifluoromethyl)phenyl)indolin-2-one as a yellow solid (1.2 g, LC/MS 85%). (LC/MS; m/z 305 [M−H]). The product was used as such without further purification.


Step 4: A solution of 3-(hydroxyimino)-1-(4-(trifluoromethyl)phenyl)indolin-2-one (200 mg, 0.65 mmol) in MeOH (5 mL) was treated with Zn dust (213 mg, 3.26 mmol) and HCl (4M in MeOH) (10 mL) at 0° C. The reaction mixture was stirred at RT for 16 h. The reaction mixture was concentrated under reduced pressure to afford 3-amino-1-(4-(trifluoromethyl)phenyl) indolin-2-one hydrochloride as a brown gum (400 mg, LC/MS 82%). (LC/MS; m/z 293 [M+H]+). The product was used as such without further purification.


Step 5: This step was executed in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to step 5 towards Cpd. No. 020. Starting material (200 mg, 0.68 mmol, LC/MS 82%) yielded crude product (110 mg, LC/MS 40%) which was purified preparative HPLC method H9. The obtained fractions were lyophilized to afford N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acrylamide (Cpd. No. 033) as a white solid (44 mg, 25%, LC/MS 99%). (LC/MS; m/z 347 [M+H]+).


The compound Cpd. No. 034 (employing Ac2O and DIPEA in DCM at step 5) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 033.


Example 7: Synthesis of 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetic acid (Cpd. No. 035)

Synthesis of methyl 2-(6-chloro-8-(1-methyl-1H-pyrazol-3-yl)imidazo[1,2-a]pyridin-2-yl)acetate (Int-2) and methyl 2-(6-chloro-8-(1-(pyridin-3-ylmethyl)-1H-pyrazol-3-yl)imidazo[1,2-a]pyridin-2-yl)acetate (Int-3)




embedded image


Step 1: A mixture of 3-bromo-5-chloropyridin-2-amine (10.0 g, 47.2 mmol) and methyl 4-chloro-3-oxobutanoate (8.5 mL, 70.9 mmol) in THF (70 mL) was heated at 66° C. for 72 h. The formed solids were filtered off, washed with THF (20 mL) and dried under reduced pressure to afford methyl 2-(8-bromo-6-chloroimidazo[1,2-a]pyridin-2-yl)acetate hydrochloride as a white solid (9.52 g, 58%). (LC/MS; m/z 303 [M+H]+). 1H NMR (400 MHz, DMSO-d6) δ ppm: 9.09 (1H, d), 8.15 (1H, s), 8.09 (1H, s), 4.01 (2H, s), 3.68 (3H, s).


Step 2: A solution of methyl 2-(8-bromo-6-chloroimidazo[1,2-a]pyridin-2-yl)acetate hydrochloride (1.50 g, 4.41 mmol), 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (964 mg, 4.63 mmol) and a 1.8M aq solution of K2CO3 (7.4 mL, 13.24 mmol) in 1,4-dioxane (22 mL) was degassed with argon for 10 min. To the solution was added Pd(dppf)Cl2 (253 mg, 0.31 mmol) and the mixture was stirred at 90° C. for 3 h. The reaction mixture was concentrated under reduced pressure, diluted with H2O (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 40 g column (silica) and a gradient of 20-100% EtOAc in n-heptane as an eluent to afford methyl 2-(6-chloro-8-(1-methyl-1H-pyrazol-3-yl)imidazo[1,2-a]pyridin-2-yl)acetate (Int-2) as a yellow solid (1.06 g, 79%). (LC/MS; m/z 305 [M+H]+).


Step 3a-b: a. A solution of methyl 2-(8-bromo-6-chloroimidazo[1,2-a]pyridin-2-yl)acetate hydrochloride (156 mg, 0.52 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (100 mg, 0.52 mmol) in 1,4-dioxane (2 mL) was degassed with argon for 10 min. To the solution was added CMBP (0.284 mL, 1.03 mmol) and stirred at 150° C. for 40 min (sealed tube; microwave irradiation). b. To the reaction mixture was added pyridin-3-ylmethanol (0.051 mL, 0.52 mmol), Pd(dppf)Cl2·DCM (42 mg, 0.052 mmol) and a 1.8M aq solution of K2CO3 (0.859 mL, 1.55 mmol). The reaction mixture was stirred at 90° C. for 2 h (sealed tube). The mixture was diluted with H2O (10 mL) and extracted with EtOAc (2×25 mL). The combined organic layer was washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 12 g column (silica) and a gradient of 0-10% MeOH in DCM as an eluent to afford methyl 2-(6-chloro-8-(1-(pyridin-3-ylmethyl)-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-2-yl)acetate (Int-3) as a brown solid (120 mg, 45%). (LC/MS; m/z 382 [M+H]+). The product contains 25% of methyl 2-(6-chloro-8-(1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-2-yl)acetate (LC/MS; m/z 291 [M+H]+). The product was used as such without further purification.


Synthesis of 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetic acid (Cpd. No. 035)



embedded image


Step 1: A solution of Int-2 (500 mg, 1.64 mmol), phenylboronic acid (240 mg, 1.97 mmol) and KF (481 mg, 8.20 mmol) in 1,4-dioxane (10 mL) and H2O (1 mL) was degassed with argon for 10 min. To the solution was added Pd(dppf)Cl2 (135 mg, 0.16 mmol) and the mixture was stirred at 140° C. for 40 min (sealed tube; microwave irradiation). The reaction mixture was diluted with H2O and extracted with EtOAc (2×100 mL). The combined organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 25 g column (silica) and a gradient of 10-100% EtOAc in n-heptane as an eluent to afford methyl 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetate as an off-white solid (200 mg, 28%, LC/MS 80%). The product was used as such without further purification. (LC/MS; m/z 347 [M+H]+).


Step 2: A solution of methyl 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetate (200 mg, 0.58 mmol) in MeOH (6 mL) and a 1M aq solution of LiOH (2.3 mL, 2.31 mmol) was stirred at RT for 1 h. The reaction mixture was concentrated under reduced pressure, dissolved in H2O (20 mL) and extracted with EtOAc (2×20 mL). To the combined organic layer was added a 1M aq solution of HCl dropwise. The formed solid were filtered off, washed with H2O and Et2O and dried under reduced pressure to afford 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetic acid (Cpd. No. 035) as a white solid (110.0 mg, 57%). (LC/MS; m/z 333 [M+H]+).


The compound Cpd. No. 036 (prepared from Int-3) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 035.


Example 8: Synthesis of 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetic acid (Cpd. No. 038)



embedded image


Step 1: A solution of Int-2 (250 mg, 0.82 mmol), K2CO3 (227 mg, 1.64 mmol), BrettPhos (22 mg, 0.041 mmol) and BrettPhos Pd G3 (38 mg, 0.041 mmol) in tBuOH (7.5 mL) was degassed with argon for 10 min. To the solution was added was added aniline (0.091 mL, 0.98 mmol) and the mixture was stirred at 110° C. for 4 h (sealed tube). The reaction mixture was diluted with H2O (25 mL) and extracted with EtOAc (2×40 mL). The combined organic layer was washed with H2O (20 mL) and brine H2O (20 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by normal phase flash column chromatography using a 12 g column (silica) and a gradient of 50-85% EtOAc in DCM as an eluent to afford methyl 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetate as a white solid (90.0 mg, 30%). (LC/MS; m/z 362 [M+H]+).


Step 2: This step was executed in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to step 2 towards Cpd. No. 035. Starting material (90 mg, 0.25 mmol) afforded 2-(8-(1-methyl-1H-pyrazol-3-yl) (phenylamino)imidazo[1,2-a]pyridin-2-yl)acetic acid (Cpd. No. 038) as a light yellow solid (74 mg, 85%). (LC/MS; m/z 348 [M+H]+).


Example 9: Synthesis of N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetamide (Cpd. No. 037)



embedded image


Step 1: To a solution of Cpd. No. 035 (50 mg, 0.15 mmol), HOAt (25 mg, 0.18 mmol), MeNH2·HCl (51 mg, 0.75 mmol) and DIPEA (0.105 mL, 0.60 mmol) in DMF (1 mL) was added EDC (0.040 mL, 0.23 mmol). The reaction mixture was stirred at RT for 16 h. The mixture was diluted with H2O (25 mL) and extracted with EtOAC (2×25 mL). The combined organic layer was washed with sat aq NH4Cl (25 mL), sat aq NaHCO3 (25 mL) and brine (15 mL), and dried over anhydrous Na2SO4. The organic layer was concentrated under reduced pressure to afford N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetamide (Cpd. No. 037) as a light brown solid (43 mg, 82%). (LC/MS; m/z 346 [M+H]+).


The compound Cpd. No. 039 (prepared from Cpd. No. 038) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 037.


Example 10: Synthesis of N-((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridin-3-yl)methyl)acrylamide (Cpd. No. 040)



embedded image


embedded image


Step-1: A solution of 1H-pyrazolo[4,3-b]pyridine (3.5 g, 29.38 mmol) in DMF (80 mL) was treated with KOH (4.94 g, 88.14 mmol) and iodine (14.914 g, 58.76 mmol) under nitrogen at 0° C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with ice water (200 mL) and extracted with EtOAc (2×200 mL). The combined organic layer was washed with ice water (100 mL), aqueous Na2S2O3 solution (100 mL), brine solution (100 mL), dried over anhydrous Na2SO4 and evaporated to afford crude compound (4.2 g) as a brown solid. The crude product was triturated with n-pentane to afford 3-iodo-1H-pyrazolo[4,3-b]pyridine (4.0 g, 56%, LCMS 98%) as a brown solid. (LCMS m/z 246 [M+H]+).


Step-2: A solution of 3-iodo-1H-pyrazolo[4,3-b]pyridine (3.0 g, 12.24 mmol) in toluene (30 mL) and water (6 mL) was treated with (4-(trifluoromethyl)phenyl)boronic acid (6.97 g, 36.73 mmol), Cu(OAC)2 (0.667 g, 3.673 mmol), K2CO3 (5.069 g, 36.73 mmol) and 1,10-Phenanthroline (0.66 g, 3.67 mmol) at room temperature. The reaction mixture was stirred under oxygen at 80° C. for 16 h. The reaction mixture was filtered through a pad of Celite and the pad was washed with EtOAc (200 mL). The filtrate was washed with water (200 mL) and extracted with EtOAc (2×100 mL). The combined organic layer was washed with brine solution (100 mL), dried over anhydrous Na2SO4 and evaporated to afford crude product as a brown semi solid (4.3 g). The crude product was purified by normal phase chromatography (CombiFlash) using 40 g column and a gradient of 20% EtOAc in petroleum ether as an eluent to afford 3-iodo-1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridine (700 mg, 15% yield) as a brown solid. (LCMS m/z 390 [M+H]+). Step-3: A solution of 3-iodo-1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridine (600 mg, 1.54 mmol) in NMP (25 mL) was treated with CuCN (483.4 mg, 5.39 mmol) at room temperature. The reaction mixture was stirred at 150° C. for 5 h. The reaction mixture was poured in cold water (100 mL) and extracted with EtOAc (2×100 mL). The combined organic layer was washed with cold water (2×50 mL), brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude product (650 mg) as a pale brown semi solid. The crude product was purified by normal phase chromatography (CombiFlash) using 40 g column and a gradient of 30% EtOAc in petroleum ether as an eluent to afford 1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridine-3-carbonitrile (250 mg, 56% yield) as a pale yellow semi solid. (LCMS m/z 289 [M+H]+).


Step-4: A solution of 1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridine-3-carbonitrile (200 mg, 0.694 mmol) in MeOH (5 mL) was treated with (Boc)2O (0.239 mL, 1.041 mmol), NiCl2·6H2O (16.5 mg, 0.069 mmol) and NaBH4 (128.4 mg, 3.5 mmol) at 0° C. and stirred at room temperature for 30 min under argon. The reaction mixture was concentrated, diluted with H2O (50 mL) and extracted with EtOAc (2×50 mL). The combined organic fraction was washed with brine (30 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude product (310 mg) as a brown semi solid, which was purified by normal phase chromatography (CombiFlash) using 40 g column using 30% EtOAc in petroleum ether as an eluent to afford tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridin-3-yl)methyl)carbamate (140 mg, 52% yield) as a pale brown solid. (LCMS m/z 393 [M+H]+).


Step-5: To a solution of tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridin-3-yl)methyl)carbamate (140 mg, 0.357 mmol) in DCM (5 mL) was added TFA (1.5 mL) at 0° C. The reaction mixture stirred at room temperature for 1 h. The resulting solution was concentrated under reduced pressure to afford crude (1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridin-3-yl)methanamine as a TFA salt (140 mg). The crude product was used in the next step without further purification. (LCMS m/z 293 [M+H]+).


Step-6: A stirred solution of (1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridin-3-yl)methanamine TFA salt (140 mg, 0.345 mmol) in 1,4 dioxane (6.0 mL) was cooled to 0° C., treated with a solution of NaHCO3 (144.82 mg, 1.724 mmol) in H2O (2.0 mL) and a solution of acryloyl chloride (34.3 mg, 0.379 mmol) in 1,4-dioxane (2.0 mL) under nitrogen. The reaction mixture was stirred at 0° C. for 30 min. The reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine solution (30 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude product as a pale brown semi solid (122 mg). The crude product was purified by reverse phase prep-HPLC and lyophilized to afford N-((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-b]pyridin-3-yl)methyl)acrylamide (Cpd. No. 040) as a white solid. (LCMS m/z 347 [M+H]+).


The compound Cpd. No. 041 (prepared from 1H-pyrazolo[4,3-c]pyridine) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to Cpd. No. 040.


Example 11: Synthesis of N-[[5-(cyanomethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]methanesulfonamide (Cpd. No. 042 with Cpd. No. 056 as an intermediate)



embedded image


embedded image


Step-1: A stirred solution of 1H-indol-5-ol (25 g, 187.76 mmol) in DMF (300 mL) was treated with potassium carbonate (38.9 g, 281.64 mmol) and benzyl bromide (26.76 mL, 225.31 mmol) at RT. The reaction mixture was stirred at 110° C. for 16 h. The progress of the reaction was monitored by TLC (Mobile phase: 20% EtOAc in pet-ether. Rf: 0.70. Detection: UV active). The reaction mixture was diluted with water (1 L) and extracted with EtOAc (2×900 mL). The combined organic layer was washed with cold water (2×500 mL), brine (500 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude product. The crude product (pre absorbed on silica) was purified by normal phase flash chromatography eluting with 8% of EtOAc in petroleum ether. The collected pure fractions were concentrated under reduced pressure to afford 5-(benzyloxy)-1H-indole (16 g, 38% yield, LCMS: 92%) as an off-white solid along with 10 g of recovered starting material. (LCMS m/z 224.3 [M+H]+).


Step-2: A solution of NaNO2 (39.55 g, 573.29 mmol) in H2O+DMF (160 mL+160 mL) was treated with drop wise addition of 2N HCl (64.0 mL) and stirred at room temperature for 30 min, followed by addition of 5-(benzyloxy)-1H-indole (16 g, 71.66 mmol) in DMF (160 mL) at 0° C. over 2 h. The reaction mixture was stirred at room temperature for 2 h. The progress of the reaction was monitored by TLC (Mobile phase: 20% EtOAc in pet-ether. Rf: 0.40. Detection: UV active). The reaction mixture was diluted with cold water (1000 mL), filtered the solid, washed with pet-ether and dried to afford crude product (13 g) as a yellow solid, which was used in the next step without any purification. (LCMS m/z 253.2 [M+H]+).


Step-3: A solution of 5-(benzyloxy)-1H-indazole-3-carbaldehyde (13 g, 51.53 mmol) in DCM (150 mL) was treated with Et3N (21.57 mL, 154.59 mmol), (4-(trifluoromethyl)phenyl)boronic acid (11.74 g, 61.83 mmol) and copper(II) acetate (18.71 g, 103.06 mmol) at RT. The reaction mixture was stirred at room temperature for 16 h under oxygen balloon. The progress of the reaction was monitored by TLC (Mobile phase: 20% EtOAc in pet-ether. Rf: 0.70. Detection: UV active). The reaction mixture was diluted with cold water (400 mL), filtered through a pad of Celite and extracted with DCM (2×1000 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (15 g, LCMS: 55%). The crude product was purified by normal phase flash chromatography (CombiFlash, 80 g column) and eluted with 8% EtOAc in pet-ether. The collected pure fractions were concentrated under reduced pressure to afford 5-(benzyloxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazole-3-carbaldehyde (4 g, LCMS: 99%) as a white solid. (LCMS m/z 397.3 [M+H]+).


Step-4: A solution of 5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazole-3-carbaldehyde (4 g, 10.09 mmol) in EtOH (60 mL) was treated with Et3N (4.22 mL, 30.27 mmol) and hydroxylamine hydrochloride (1.40 mg, 20.18 mmol) at RT. The reaction mixture was stirred at 80° C. for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.40. Detection: UV active). The reaction mixture was diluted with cold water (500 mL) and extracted with EtOAc (2×500 mL). The combined organic layer was washed with brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (4 g, LCMS: 96%) as a yellow solid, which was used in the next step without purification. (LCMS m/z 412.6 [M+H]+).


Step-5: A stirred solution of 5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazole carbaldehyde oxime (4 g, 9.72 mmol) in Et2O (100 mL) was treated with drop wise addition of 2M LAH in THF solution (9.72 mL, 19.44 mmol) at 0° C. The reaction mixture was stirred at room temperature for 3 h, progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.10. Detection: UV active). The reaction mixture was quenched with saturated sodium sulphate solution (2.0 mL) and EtOAc (1000 mL). The mixture was stirred for 30 min and filtered. The organic layer was dried over Na2SO4 and concentrated under reduced pressure to afford crude product (3 g, LCMS 72%). The crude product was purified by chromatography (CombiFlash, 40 g silica column) and eluted with 5% MeOH in DCM. The collected pure fractions were concentrated under reduced pressure to afford 5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine (2.5 g, LCMS: 80%) as a white solid. (LCMS m/z 398.4 [M+H]+).


Step-6: A solution of (5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine (500 mg, 1.26 mmol) in DCM (10 mL) was treated with Et3N (0.53 mL, 3.77 mmol), followed by MsCl (0.117 mL, 1.51 mmol) at 0° C. and the reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.81. Detection: UV active). After consumption of starting material on TLC, the reaction mixture was diluted with water (100 mL), and extracted with DCM (2×75 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered & concentrated under reduced pressure to afford crude product (500 mg). The crude product (pre absorbed on silica) was purified by normal phase flash chromatography (Grace Instrument, 24 g silica column) and eluted with 20% of EtOAc in pet-ether gradient. The collected pure fractions were concentrated under reduced pressure to afford N-((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl) methanesulfonamide (Cpd. No. 056, 300 mg, 50% yield) as a brown solid. (LCMS m/z 276.3 [M+H]+).


Step-7: To a solution of N-((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide (300 mg, 0.63 mmol) in DCM (6 mL) was added 1 M BBr3 in DCM (1.89 mL, 1.89 mmol), at 0° C., then the reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.56. Detection: UV active). After consumption of starting on TLC, the reaction mixture was diluted with water (40 mL) and extracted with EtOAc (2×40 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude N-((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide (240 mg, LCMS: 85%) as a brown solid, which was used in the next step without any purification. (LCMS m/z 386.3 [M+H]+).


Step-8: A solution of N-((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide (150 mg, 0.39 mmol) in ACN (3 mL) was treated with K2CO3 (107.6 mg, 0.78 mmol) and 2-chloroacetonitrile (45.2 mg, 0.58 mmol) at room temperature. The reaction mixture was stirred at room temperature for 24 h. Progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.50. Detection: UV active). After consumption of starting on TLC, the reaction mixture was diluted with water (30 mL) and extracted with EtOAc (2×40 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, and concentrated under reduced pressure to afford crude product (150 mg). The crude compound was purified by Prep-HPLC Method H2. The collected pure fractions were concentrated and lyophilized to afford N-((5-(cyanomethoxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide (Cpd. No. 042, 20 mg, 12% yield) as a white solid.


Cpd. No. 054 (prepared from ((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Step-5 of the preparation of Cpd. No. 43.


Cpd. No. 055 (prepared from ((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Step-2 of Example 16.


Example 12: Synthesis of N-[[5-(cyanomethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide (Cpd. No. 043)



embedded image


embedded image


Step-1: A stirred solution of ((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine (2.5 g, 2.02 mmol) in THF+MeOH (37.0 mL+12.0 mL) was treated with drop wise addition of Et3N (1.31 mL, 9.43 mmol) and Boc-anhydride (1.73 mL, 7.54 mmol) at 0° C. The reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.70. Detection: UV active). The reaction mixture was diluted with cold water (100 mL) and extracted with EtOAc (2×500 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (3.0 g). The crude product was purified by normal phase flash chromatography (CombiFlash, 24 g column) and eluted with 8% EtOAc in pet-ether. The collected pure fractions were concentrated under reduced pressure to afford tert-butyl-((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (2.5 g, LCMS: 90%) as a white solid. (LCMS m/z 498.2 [M+H]+).


Step-2: A solution of tert-butyl-((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (1.8 g, 3.62 mmol) in MeOH (30 mL) was treated with 10% Pd/C (1.9 g, 18.09 mmol) and ammonium formate (2.28 g, 36.18 mmol) at RT. The reaction mixture was stirred at 70° C. for 2 h. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.35. Detection: UV active). The reaction mixture was filtered through a pad of Celite and the pad was washed with MeOH (150 mL). The filtrate was concentrated under reduced pressure, diluted with water (200 mL) and extracted with EtOAc (2×200 mL). The combined organic layers were washed with brine (150 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (1.8 g, LCMS: 94%). The crude compound was triturated with pet-ether (2×10 mL) and dried to afford tert-butyl-((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (1.2 g, LCMS: 97%) as a white solid. (LCMS m/z 384.35 [M+H]+).


Step-3: A solution of tert-butyl ((5-hydroxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl)carbamate (150 mg, 0.37 mmol) in DMF (10 mL) was treated with K2CO3 (254.4 mg, 1.84 mmol) and 2-bromoacetonitrile (131.3 mg, 1.10 mmol) at room temperature. The reaction mixture was stirred at room temperature for 12 h, monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.58. Detection: UV). The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (2×50 mL). The organic layer was washed with brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (150 mg, LCMS: 90%). The crude product (pre absorbed on silica) was purified by normal phase flash chromatography (Grace Instrument) using 24 g column cartridge and eluted with 70% of EtOAc in pet-ether. The collected pure fractions were concentrated under reduced pressure to afford tert-butyl ((5-(cyanomethoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl)methyl)carbamate (120 mg, 73% yield) as a white solid. (LCMS m/z 447.2 [M+H]+).


Step-4: A solution of tert-butyl ((5-(cyanomethoxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (120 mg, 0.27 mmol) in DCM (5 mL) was treated with TFA (0.21 mL, 2.68 mmol) at 0° C. The reaction mixture was stirred at room temperature for 2 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.15. Detection: UV active). The reaction mixture was concentrated under reduced pressure and dried to afford crude 2-((3-(aminomethyl)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-5-yl)oxy)acetonitrile hydrochloride (120 mg, LCMS: 49%) as a brown solid. The crude product was used in the next step without purification. (LCMS m/z 347.3[M+H]+).


Step-5: A solution of 2-((3-(aminomethyl)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-5-yl)oxy)acetonitrile hydrochloride (LCMS: 49%) (120 mg, 0.31 mmol) in 1,4-dioxane+H2O (3 mL+1 mL) was treated with sodium bicarbonate (131.68 mg, 1.57 mmol) and acryloyl chloride (31.2 mg, 0.34 mmol) at 0° C. The reaction mixture was stirred at room temperature for 30 min. The progress of the reaction was monitored by TLC (Mobile phase: 70% EtOAc in pet ether. Rf: 0.75. Detection: UV active). After consumption of starting on TLC, the reaction mixture was diluted with water (30 mL), extracted with EtOAc (2×30 mL). The combined organic layers were washed with brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (120 mg, LCMS: 49%), which was purified by Prep HPLC Method H2. The collected pure fractions were concentrated and lyophilized to afford N-((5-(cyanomethoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl)methyl)acrylamide (Cpd. No. 043, 40 mg, 12% yield) as a white solid.


Cpd. No. 044 (prepared from tetrahydrofuran-3-yl)methyl 4-methylbenzenesulfonate and tert-butyl ((5-hydroxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl)carbamate) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used for preparation of Cpd. Nos. 042 and 043.


Cpd. No. 046 (prepared from (5-((tetrahydrofuran-3-yl)methoxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine hydrochloride) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Step-5 of the preparation of Cpd. No. 043. This racemate was purified by chiral prep-SFC (Chiralcel OX-H (250×4.6×5μ) % CO2: 85%, % Co solvent: 15% (100% MeOH). Total flow: 3.0 g/min. Back Pressure: 100.0 bar. Temperature: 30.0° C., UV: 215 nm. Stack time: 8.0 min.). Collected fractions were concentrated and lyophilized to afford Cpd. No. 047 (21.78 mg) as an off-white sticky solid and Cpd. No. 048 (18.97 mg) as an off-white sticky solid. The absolute configurations of the isolated enantiomers were not determined.


Cpd. Nos. 063, 064, 065, 066, 067 and 068 were prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd No. 043.


Example 13: Synthesis of N-[[5-phenoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]methanesulfonamide (Cpd. No. 45)



embedded image


Step-1: A solution of tert-butyl ((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (500 mg, 1.22 mmol) in DMSO (12 mL) was treated with Iodobenzene (300.4 mg, 1.47 mmol) and K3PO4 (781.5 mg, 3.68 mmol) at RT. The reaction mixture was degassed for 5 minutes with N2. Picolinic acid (30.2 mg, 0.24 mmol) and copper iodide (23.37 mg, 0.24 mmol) were added and again the reaction was degassed for 5 minutes. The reaction mixture was stirred at 120° C. for 1 h in microwave. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.81. Detection: UV active). The reaction mixture was diluted with cold water (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with cold water (25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (500 mg, LCMS: 82%). The crude compound (pre absorbed on silica) was purified by normal phase flash chromatography (Grace Instrument) using 40 g column cartridge and eluted with 10% of EtOAc in pet-ether. The collected pure fractions were concentrated under reduced pressure to afford tert-butyl ((5-phenoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (400 mg, 67% yield) as a white solid. (LCMS m/z 484.4 [M+H]+).


Step-2: A solution of tert-butyl ((5-phenoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (400 mg, 0.83 mmol) in DCM (5 mL) was treated with 4M HCl in dioxane (3.3 mL, 13.24 mmol) at 0° C. and the reaction mixture was stirred at room temperature for 2 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.12. Detection: UV active). The reaction mixture was concentrated under reduced pressure and dried to afford crude product (350 mg, LCMS: 95.24%) as a white solid, which was used in the next step without purification. (LCMS m/z 384.4 [M+H]+).


Step-3: A solution of (5-phenoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine hydrochloride (180 mg, 0.43 mmol) in DCM (5 mL) was treated with triethylamine (0.3 mL, 2.14 mmol) and methane sulfonyl chloride (0.04 mL, 0.51 mmol) at 0° C. The reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.73. Detection: UV active). The reaction mixture was diluted with water (20 mL) and extracted with DCM (2×30 mL). The combined organic layer was washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (200 mg, LCMS: 84.75%), which was purified by prep-HPLC Method H2. Collected pure fractions were concentrated and lyophilized to afford N-((5-phenoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide (Cpd. No. 045, 120 mg, 60% yield) as an off-white solid.


Cpd. No. 049 (prepared from (5-phenoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine hydrochloride) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Step-5 of the preparation of Cpd No. 043.


Cpd. No. 051 (prepared from tert-butyl ((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate and 6-chloro-1-methylpyrimidine-2,4(1H,3H)-dione) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Steps-1 and 2 of the preparation of Cpd. No. 045 and Step-5 of the preparation of Cpd. No. 043.


Example 14: Synthesis of N-[[6-(cyclopropoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide (Cpd. No. 050)



embedded image


embedded image


Step-1: A solution of NaNO2 (101.63 g, 1195.83 mmol) in water (175 mL) and DMF (175 mL) was cooled to 0° C., treated with 3N HCl (150 mL, 448.44 mmol) at 0° C. and stirred for 20 min at 0° C. A solution of 6-methoxy-1H-indole 1 (22 g, 149.48 mmol) in DMF (85 mL) was added to the reaction mixture at 0° C. over period of 1 h and stirred at RT for 4 h. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in Pet-ether; Detection: UV-active; Rf: 0.22). The reaction mixture was diluted with water (200 mL) and extracted with EtOAc (3×150 mL). The combined organic layer was washed with brine solution (2×300 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude product (30 g, LCMS 37.5%) as a black gum, which was purified by normal phase chromatography (Grace) using 120 g column and EtOAc in Pet-ether to afford 6-methoxy-1H-indazole-3-carbaldehyde (6 g; LCMS purity 88.2%) as an off-white solid. (LCMS m/z 177.3 [M+H]+).


Step-2: A solution of 6-methoxy-1H-indazole-3-carbaldehyde (10 g, 56.76 mmol) in 1,4-dioxane (250 mL) was treated with 1-iodo-4-(trifluoromethyl)benzene (18.52 g, 68.11 mmol), K2CO3 (19.61 g, 141.90 mmol) at RT. The reaction mixture was degassed with argon for 5 min, treated with trans-N, N′-dimethylcyclohexane-1,2-diamine (1.04 g, 7.37 mmol) and CuI (1.29 g, 6.81 mmol) at RT. The reaction mixture was stirred at 80° C. for 16 h. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.35. Detection: UV active). The reaction mixture was filtered through a pad of Celite, washed with EtOAc (200 mL). The filtrate was washed with water (150 mL), brine (100 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude product as a brown solid (12 g, LCMS purity 66.7%), which was purified by flash column chromatography (CombiFlash) using 40 g column and 18% EtOAc in pet ether to afford 6-methoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazole-3-carbaldehyde as a yellow solid (4 g; LCMS purity 81.68%). (LCMS m/z 321.3 [M+H]+).


Step-3: A solution of 6-methoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazole-3-carbaldehyde (4 g, 12.48 mmol) in MeOH (100 mL) was treated with NaBH4 (0.70 g, 18.73 mmol) at 0° C. and slowly warmed to RT over 2 h. Progress was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.13. Detection: UV active). The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (2×150 mL). The combined organic layer was washed with brine solution (150 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude (6-methoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanol (3.7 g; LCMS purity 94%) as a yellow solid. The compound was used in the next step without further purification. (LCMS m/z 323.4 [M+H]+).


Step-4: A solution of (6-methoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanol (3.70 g, 11.48 mmol) in THF (50 mL) was treated with isoindoline-1,3-dione (2.53 g, 17.22 mmol), diisopropyl azodicarboxylate (6.96 g, 34.44 mmol) and PPh3 (9.03 g, 34.44 mmol) at 0° C. The reaction mixture was stirred at RT for 5 h, progress of the reaction was monitored by LCMS and TLC (Mobile phase: 30% EtOAc in Pet-ether. Detection: UV-Active. Rf: 0.30). The reaction was diluted with water (40 mL) and extracted with EtOAc (3×50 mL). The combined organic layer was washed with brine solution (50 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude product as a brown gum (5.0 g, LCMS purity 52%), which was purified by reverse phase chromatography (Grace) using C18 column and a gradient of CAN in water to afford 2-((6-methoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)isoindoline-1,3-dione as an off-white solid (3.0 g; LCMS purity 97%). (LCMS m/z 452.4 [M+H]+).


Step-5: A solution of 2-((6-methoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)isoindoline-1,3-dione (2.8 g, 6.20 mmol) in DCM (36 mL) was treated with BBr3 (1.0 M in DCM) (31 mL, 31.01 mmol) at 0° C. The reaction mixture was stirred at RT for 16 h, progress of the reaction was monitored by TLC (Mobile phase: 50% EtOAc in Pet-ether. Detection: UV-Active. Rf: 0.25). The reaction was quenched with MeOH (40 mL) and saturated NaHCO3 solution (40 mL) at 0° C. and extracted with DCM (2×200 mL). The combined organic layer was washed with brine (2×250 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude residue, which was triturated with diethyl ether (15 mL) and dried under high vacuum to afford 2-((6-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)isoindoline-1,3-dione (1.8 g, LCMS purity 95%) as a yellow solid. (LCMS m/z 438.3 [M+H]+).


Step-6: A solution of 2-((6-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)isoindoline-1,3-dione (1.9 g, 4.34 mmol) in toluene (32 mL) and water (8 mL) was treated with copper(II) acetate (316 mg, 1.73 mmol), 1,10-phenanthroline (313 mg, 1.73 mmol) and potassium cyclopropyltrifluoroborate (2.57 g, 17.37 mmol) at RT under oxygen balloon. The reaction mixture was stirred at 70° C. for 24 h. Progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in Pet ether. Rf: 0.4. Detection: UV active). The reaction mixture was cooled to RT, diluted with aqueous NH4Cl solution (200 mL) and extracted with EtOAc (2×250 mL). The combined organic layer was washed with brine (2×150 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude product (LCMS 32% of desired mass+18% of starting material) as a black gum, which was purified by normal phase column chromatography and 15% EtOAc in Pet ether as an eluent to afford 2-((6-cyclopropoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)isoindoline-1,3-dione (320 mg; LCMS purity 81.51%) as an off-white solid. (LCMS m/z 478.3 [M+H]+).


Step-7: A solution of 2-((6-cyclopropoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)isoindoline-1,3-dione (320 mg, 0.67 mmol) in EtOH (5 mL) was treated with N2H4·H2O (0.16 mL, 3.35 mmol) at RT. The reaction mixture was stirred at 85° C. for 2 h, monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.19. Detection: UV active). The reaction mixture was cooled to RT, filtered and the filtrate was concentrated under reduced pressure. The resulting residue was diluted with EtOAc (50 mL) washed with brine (2×25 mL), dried over sodium sulphate and concentrated under reduced pressure to afford crude product (190 mg; LCMS purity 77.5%) as a yellow solid. (LCMS m/z 348.2 [M+H]+).


Step-8: A solution of crude (6-cyclopropoxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methoxamine (15 mg, 0.043 mmol) in 1,4-dioxane (5 mL) was treated with NaHCO3 (18.15 mg, 0.21 mmol) and acryloyl chloride (4.69 mg, 0.052 mmol) in dioxane (1.0 mL) at 0° C. The reaction mixture was stirred at 0° C. for 30 min, progress of the reaction was monitored by TLC (mobile phase: 10% MeOH in DCM. Rf: 0.51. Detection: UV active). The reaction mixture was diluted with EtOAc (15 mL), washed with Water (2×10 mL), the combined organic layer was dried over Na2SO4 concentrated under reduced pressure to afford crude (30 mg, LCMS 50%) as a brown gum. The crude product was purified by Prep-HPLC using Method H13 and obtained fractions were concentrated under reduced pressure to afford N-((6-cyclopropoxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) acrylamide (3.20 mg, LCMS 99.6%, Yield: 1.7%) as an off-white solid.


Cpd. No. 061 (prepared from (6-cyclopropoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Step-6 of the preparation of Cpd. No. 042.


Cpd. No. 062 (prepared from (6-cyclopropoxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine and propionyl chloride) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used for the preparation of Cpd. No. 050.


Example 15: Synthesis of N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]methanesulfonamide (Cpd. No. 052)



embedded image


embedded image


Step-1: A solution of 1H-pyrazole-5-carbaldehyde (4 g, 41.62 mmol) in DCM (80 mL) was treated with pTSA (716.00 mg, 4.16 mmol) followed by the addition of DHP (7.53 mL, 83.25 mmol). The reaction mixture was stirred at room temperature for 12 h. The reaction was monitored by TLC (Mobile phase: 50% EtOAc in pet ether. Rf: 0.75. Detection: UV active). The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (2×100 mL). The combined organic layer was washed with brine solution (100 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product as a brown gum (5 g, LCMS 60%). The crude product was purified by normal phase column chromatography (combi) by using a 48 g column and a gradient of 0-50% EtOAc in pet ether as an eluent to afford 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-5-carbaldehyde (3.5 g, LCMS 94.8%) as pale-yellow liquid. (LCMS m/z 181.3 [M+H]+).


Step-2: To a stirred solution of 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carbaldehyde (3.5 g, 19.44 mmol) in MeOH (40 mL) was added NaBH4 (1.43 g, 38.88 mmol) portion wise at 0° C. The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was monitored by TLC (Mobile phase: 50% EtOAc in Pet ether. Rf: 0.15. Detection: UV active, charring with KMnO4). The reaction mixture was concentrated, diluted with water (100 mL) and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine solution (100 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product (4 g, LCMS 80%). The crude product was purified by normal phase chromatography using a 40 g column and the obtained pure fractions were concentrated under reduced pressure to afford (1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methanol (2.7 g, LCMS 92%) as a white liquid. (LCMS m/z 183.21[M+H]+).


Step-3: A stirred solution of (1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methanol (1 g, 5.48 mmol) in DCM (10 mL) was treated with TPP (1.43 g, 5.48 mmol) followed by the addition of CBr4 (2.18 g, 6.58 mmol) at 0° C. The reaction was stirred at room temperature for 16 h. The reaction mixture was monitored by TLC (Mobile phase: 50% EtOAc in Pet ether. Rf: 0.77. Detection: UV Active). The reaction mixture was diluted with water (70 mL) and extracted with EtOAc (2×70 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure to obtained crude product (1 g, LCMS 25%) as a brown gum. The crude product was purified by Combi Flash (normal phase) using a 40 g column and 20% EtOAc in pet ether as an eluent to afford 3-(bromomethyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (700 mg, LCMS 93.9%) as a white solid. (LCMS m/z 245 [M+H]+).


Step-4: A stirred solution of N-((5-hydroxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) methanesulfonamide (from Example 11, 250 mg, 0.64 mmol) in THF (3 mL) was treated with 3-(bromomethyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (191.29 mg, 0.77 mmol) followed by K2003 (134.41 mg, 0.97 mmol) at room temperature. The reaction mixture was stirred at room temperature for 3 days. The reaction mixture was monitored by TLC (Mobile phase: 50% EtOAc in Pet ether, Rf: 0.31. Detection: UV). The reaction mixture was diluted water (30 mL) and extracted with EtOAc (3×25 mL). The combined organic layer was washed with brine solution (25 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product (200 mg, LCMS 42%). The crude product was purified by normal phase flash column chromatography (Combi Flash, 48 g silica gel column) eluted with 40% EtOAc in pet ether to afford N-((5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)methoxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide (180 mg, LCMS 62%) as a white solid. (LCMS m/z: 550 [M+H]+).


Step-5: A stirred solution of N-((5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) methanesulfonamide (180 mg, 0.32 mmol) in DCM (3 mL) was treated with TFA (1 mL) at 0° C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was monitored by TLC (Mobile phase: 10% Methanol in DCM. Rf: 0.29. Detection: UV active). The reaction mixture was concentrated under reduced pressure and dried. The residue was triturated with diethyl ether (2×10 mL) and dried under high vacuum to afford crude product (150 mg, LCMS 53%) as a brown gum. The crude compound was purified by prep-HPLC Method H2 and the fractions were concentrated under reduced pressure to afford (N-((5-((1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) methanesulfonamide (Cpd. No. 052) (37.9 mg, LCMS 95.0%) as a white solid.


Example 16: Synthesis of N-[[5-(3-methyl-2,6-dioxo-pyrimidin-4-yl)oxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]propenamide (Cpd. No. 053)



embedded image


Step-1: A solution of tert-butyl ((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)carbamate (from Example 12, 200 mg, 0.49 mmol) in DCM (5 mL) was treated with 4 M HCl in dioxane (1.22 mL) at 0° C. The reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.1. Detection: UV active). The reaction mixture was concentrated under reduced pressure to afford crude product (160 mg, LCMS: 68%), which was used in the next step without further purification. (LCMS m/z 308.37 [M+H]+).


Step-2: A solution of 3-(aminomethyl)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-5-ol hydrochloride (160 mg, 0.46 mmol) in 1,4-dioxane+H2O (3 mL+1 mL) was treated with sodium bicarbonate (195.5 mg, 2.33 mmol) followed by propionyl chloride (172.8 mg, 1.86 mmol) at 0° C. The reaction mixture was stirred at room temperature for 3 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.59. Detection: UV active). The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4 & concentrated under reduced pressure to afford crude (160 mg, LCMS: 61%) as a brown gum. The crude product was purified by normal phase flash chromatography (Grace Instrument) and eluted with 50% EtOAc in pet-ether. The collected pure fractions were concentrated under reduced pressure to afford N-((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)propionamide (95 mg, LCMS: 90.79%) as a brown solid. (LCMS m/z 488.2 [M+H]+).


Step-3: This step was carried out as in Step-1 of Example 13, starting with N-((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)propionamide (85 mg, 0.23 mmol) and 6-chloro-1-methylpyrimidine-2,4(1H,3H)-dione (45.07 mg, 0.28 mmol). The crude product was purified by prep-HPLC Method H15 and the collected pure fractions were concentrated under reduced pressure to afford N-((5-((3-methyl-2,6-dioxo-1,2,3,6-tetrahydropyrimidin-4-yl) oxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) propionamide (Cpd. No. 053, 16.3 mg, LCMS 99.8%) as a white solid.


Cpd. No. 060 (prepared from N-((5-hydroxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) methanesulfonamide (see Example 11)) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in Step-3 of the preparation of Cpd. No. 053.


Example 17: Synthesis of N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide (Cpd. No. 058)



embedded image


embedded image


Step-1: A solution of (5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methanamine (1 g, 2.52 mmol) in DCM (10 mL) was treated with trimethylamine (1.06 mL, 7.55 mmol), followed by trifluoroacetic anhydride (0.42 mL, 3.02 mmol) at 0° C. The reaction mixture was stirred at room temperature for 2 h. The progress of the reaction was monitored by TLC (Mobile phase: 10% MeOH in DCM. Rf: 0.77. Detection: UV active). The reaction mixture was concentrated under reduced pressure, then diluted with water (100 mL) and extracted with EtOAc (2×100 mL). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, filtered & concentrated under reduced pressure to afford crude N-((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)-2,2,2-trifluoroacetamide (1.2 g, LCMS: 89%) as a brown solid. The crude product was used in the next step without further purification. (LCMS m/z: 494.31 [M+H]+).


Step-2: A solution of N-((5-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)-2,2,2-trifluoroacetamide (1.2 g, 2.43 mmol) in methanol (15 mL) was treated with palladium on activated carbon (1.29 g, 12.16 mmol), followed by ammonium formate (1.53 g, 24.32 mmol) at room temperature. The reaction mixture was stirred at 80° C. for 1 h. The progress of the reaction was monitored by TLC (Mobile phase: 30% EtOAc in pet-ether. Rf: 0.25. Detection: UV active). The reaction mixture was filtered through a celite pad with MeOH (100 mL). The filtrate was dried over Na2SO4 and concentrated under reduced pressure to afford crude product (1 g, LCMS: 75%). The crude product was purified by normal phase chromatography (Grace Instrument) using a 40 g column eluting with 30% EtOAc in pet ether (250 mL) to afford 2,2,2-trifluoro-N-((5-hydroxy-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acetamide (650 mg, LCMS: 98%, 66% yield) as an off white solid. (LCMS m/z: 404.30 [M+H]+).


Step-3: A solution of 2,2,2-trifluoro-N-((5-hydroxy-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) acetamide (650 mg, 1.61 mmol) in ACN (7 mL) was treated with 3-(bromomethyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (472.25 mg, 1.93 mmol) followed by the addition of K2CO3 (267.09 mg, 1.93 mmol) at room temperature. The reaction mixture was stirred at room temperature for 3 days. The reaction was monitored by TLC (Mobile phase: 30% EtOAc in Pet ether. Rf: 0.41. Detection: UV active) and completion of the reaction was confirmed by LCMS. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude compound (700 mg, LCMS 73%) as a brown gum, which was purified by column chromatography (100-200 silica gel) eluted with 50% EtOAc in Pet ether to afford 2,2,2-trifluoro-N-((5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)methoxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acetamide (500 mg, LCMS purity 91%) as a white solid. (LCMS m/z: 568.57 [M+H]+).


Step-4: A solution of 2,2,2-trifluoro-N-((5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) acetamide (530 mg, 0.106 mmol) in MeOH (5 mL) was treated with 1N Aq NaOH solution (2 mL) at room temperature. The reaction mixture was stirred at RT for 16 h, monitored by TLC (Mobile phase: 5% MeOH in DCM. Rf: 0.42. Detection: UV active) and completion of the reaction was confirmed by LCMS. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford (5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methanamine (450 mg, LCMS: 88%) as a brown solid. The crude compound was used as such for the next step without further purification. (LCMS m/z: 472.17 [M+H]+).


Step-5: A solution of (5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methanamine (250 mg, 0.53 mmol) in 1,4-dioxane+H2O (5.0 mL+1.8 mL) was treated with NaHCO3 (111.46 mg, 1.327 mmol) and acryloyl chloride (52.84 mg, 0.58 mmol) at 0° C. The reaction mixture was stirred for at room temperature for 2 h, monitored by TLC (Mobile phase: 5% MeOH in DCM. Rf: 0.56. Detection: UV active). The reaction mixture was diluted with EtOAc (30 mL), washed with brine (2×50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford N-((5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) acrylamide (250 mg, LCMS 82%) as a brown gum. The compound was used as such for the next step without further purification. (LCMS m/z: 526.40 [M+H]+).


Step-6: A solution of N-((5-((1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl) methoxy)-1-(4-(trifluoromethyl) phenyl)-1H-indazol-3-yl) methyl) acrylamide (250 mg, 0.47 mmol) in DCM (4.0 mL) was treated with TFA (1 mL) at 0° C. The reaction mixture was stirred at room temperature for 2 h, monitored by TLC (Mobile phase: 70% EtOAc in pet ether. Rf: 0.29. Detection: UV active). The reaction mixture was concentrated under reduced pressure to afford crude compound (200 mg, LCMS 74%) as a brown gum. The crude compound was purified by Prep-HPLC Method H5 and pure fractions were concentrated under reduced pressure to afford N-((5-((1H-pyrazol-3-yl)methoxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acrylamide (Cpd. No. 058, 69.8 mg, yield 33%, LCMS 99.4%) as a white solid.


Example 18: Synthesis of N-[[1-[4-(trifluoromethyl)phenyl]pyrazolo[3,4-c]pyridin-3-yl]methyl]prop-2-enamide (Cpd. No. 059)



embedded image


Step-1: A solution of 3-bromo-1H-pyrazolo[3,4-c]pyridine (2.0 g, 10.10 mmol) in toluene (30 mL) and H2O (6 mL) was treated with (4-(trifluoromethyl)phenyl)boronic acid (5.7 g, 30.30 mmol), K2CO3 (4.18 g, 30.3 mmol), Cu(OAc)2 (0.54 g, 30.03 mmol) and 1,10-phenanthroline (0.54 g, 30.03 mmol) under O2 balloon at room temperature. The reaction mixture was stirred at 80° C. for 16 h, monitored by TLC (mobile phase: 20% EtOAc in pet ether. Rf: 0.35·TLC detection: UV active). The resulting solution was diluted with H2O (100 mL) and extracted with EtOAc (2×60 mL). The combined organic layer was washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude compound (2.0 g, LCMS 34%). The crude compound was purified by normal phase flash column chromatography (CombiFlash, 80 g silica gel column) and eluted with 30% EtOAc in pet ether to afford 3-bromo-1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[3,4-c]pyridine (470 mg, 13%) as a pale yellow solid. (LCMS m/z 342.08[M+H]+).


Step-2: A solution of 3-bromo-1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[3,4-c]pyridine (470 mg, 1.38 mmol), tert-butyl ((trifluoro-l4-boraneyl)methyl)carbamate, potassium salt (393.1 mg, 1.65 mmol) and K3PO4 (879 mg, 4.14 mmol) in t-BuOH (5 mL) and H2O (5 mL) was degassed with argon at room temperature for 5 min. To this mixture was added s-Phos (56.6 mg, mg, 0.13 mmol) and Pd(OAc)2 (30.9 mg, 0.13 mmol) at room temperature. The reaction mixture was stirred at 70° C. for 16 h in a sealed tube, monitored by TLC (mobile phase: 50% EtOAc in Pet ether, Rf=0.4, TLC detection: UV). The resulting solution was diluted with H2O (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was dried with anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product (450 mg, LCMS 24%). The crude product was purified by flash column chromatography (12 g CombiFlash), eluted with 25% EtOAc in Pet Ether and collected pure fractions were concentrated under vacuum to afford tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[3,4-c]pyridin-3-yl)methyl)carbamate (120 mg, 23%) as an off white solid. (LCMS m/z 393.34 [M+H]+).


Step-3: A solution of tert-butyl ((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[3,4-c]pyridin-3-yl)methyl)carbamate (120 mg, 0.30 mmol) in DCM (3 mL) was treated with TFA (1 mL) at 0° C. The reaction mixture was stirred at room temperature for 2 h, monitored by TLC (mobile phase: 10% MeOH in DCM. Rf: 0.1, TLC detection: UV-active). The reaction mixture was concentrated under reduced pressure, triturated with diethyl ether (2×5 mL) and dried under vacuum to afford crude compound (90 mg, LCMS 92%). The crude product was use for next step without further purification. (LCMS m/z: 293.29 [M+H]+).


Step-4: A solution of (1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[3,4-c]pyridin-3-yl)methanamine·TFA salt (90 mg, 0.22 mmol) in DCM (4 mL) was treated with TEA (134 mg, 1.33 mmol) and a solution of acryloyl chloride (20 mg, 0.22 mmol) in DCM (1 mL) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at 0° C. for 30 min, monitored by TLC (mobile phase: 10% MeOH in DCM. Rf: 0.4. detection: UV active). The resulting solution was diluted with H2O (30 mL) and extracted with DCM (50 mL). The organic fraction was dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product (80 mg, LCMS 62%). The crude product was purified by prep-HPLC Method H13 and collected fractions were concentrated under reduced pressure to remove organic solvents and the aqueous layer was lyophilized to afford N-((1-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[3,4-c]pyridin-3-yl)methyl)acrylamide (Cpd. No. 059, 27.2 mg, 38%) as an off-white solid.


Cpd. No. 071 (prepared from 3-bromo-5-(trifluoromethyl)-1H-indazole [1086378-32-6]) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd. No. 059.


Cpd. No. 072 was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd. No. 071, except that acetyl chloride was used in the final step.


Cpd. No. 073 was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd. No. 071, except that methanesulfonyl chloride was used in the final step.


Cpd. No. 074 (prepared from 3-Bromo-6-(trifluoromethyl)-1H-indazole [1000341-21-8]) was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd. No. 059.


Cpd. No. 075 was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd. No. 074, except that acetyl chloride was used in the final step.


Cpd. No. 076 was prepared in a manner similar (use of appropriate reagents and purification methods known to the person skilled in the art) to that used in the preparation of Cpd. No. 074, except that methanesulfonyl chloride was used in the final step.


Example 19: Synthesis of 2-[1-[4-(trifluoromethyl)phenyl]indazol-4-yl]oxyacetic acid (Cpd. No. 069)



embedded image


Step-1: A stirred solution of 4-(benzyloxy)-1H-indazole (500 mg, 2.23 mmol) in DCM (50 mL) was treated with (4-(trifluoromethyl)phenyl)boronic acid (847 mg, 4.46 mmol), Cu(OAc)2 (607.4 mg, 3.34 mmol), pyridine (352.7 mg, 4.46 mmol) and 4 Å molecular sieves powder (500 mg) at room temperature under air balloon. The reaction mixture was stirred at room temperature for 18 h under air balloon, monitored by TLC (Mobile phase: 30% EtOAc in Hexane, Rf: 0.69, TLC detection: UV). Reaction mixture was filtered on Celite bed and washed with EtOAc (50 mL), filtrate was concentrated and diluted with water (30 mL) and extracted in EtOAc (2×30 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to get crude compound (800 mg, LCMS: 58%+12% regio-isomers and 10% SM) as blue coloured liquid. The crude compound was purified by normal phase column chromatography (Combi, 100-120 mess silica size) with a gradient of 5%-6% EtOAc in pet ether as an eluent. The combined pure fractions were collected and evaporated under reduced pressure to afford 4-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazole (320 mg, LCMS: 88%+9% regio-isomers) as an pale yellow solid. (LCMS m/z: 369.54 [M+H]+).


Step-2: A stirred solution of 4-(benzyloxy)-1-(4-(trifluoromethyl)phenyl)-1H-indazole (300 mg, 0.814 mmol) in MeOH (8 mL) was treated with 10% Pd/C (150 mg, 50% moisture) at room temperature. Reaction mixture was stirred at room temperature under hydrogen atmosphere for 5 h, monitored by TLC (Mobile phase: 10% EtOAc in Hexane, Rf: 0.11, TLC detection: UV). The reaction mixture was filtered on Celite bed and washed the bed with MeOH (2×40 mL). The filtrated was concentrated under reduced pressure to get crude product 1-(4-(trifluoromethyl)phenyl)-1H-indazol-4-ol (220 mg, LCMS: 92%) as an off white solid. (LCMS m/z: 279.50 [M+H]+).


Step-3: A stirred solution of 1-(4-(trifluoromethyl)phenyl)-1H-indazol-4-ol (190 mg, 0.683 mmol) in DMF (4.0 mL) was treated with tert-butyl 2-(bromomethyl)acrylate (160 mg, 0.82 mmol) and K2CO3 (266.5 mg, 1.36 mmol) at room temperature. The reaction mixture was heated to 80° C. and stirred for 3 h, monitored by TLC (Mobile phase: 20% EtOAc in Hexane, Rf: 0.4, TLC detection: UV). The reaction mixture was diluted with cold water (15 mL) and extracted in cold EtOAc (2×25 mL). The combined organic layer was dried over Na2SO4 and concentrated under reduced pressure to get crude product (290 mg, LCMS: 91%) as a brown liquid. The crude compound was purified by Combi (100-200 mess silica, 40 g cartridge) using 5-6% EtOAc in pet ether as an eluent. Pure fractions were collected and concentrated under reduced pressure to get pure compound tert-butyl 2-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-4-yl)oxy)acetate (220 mg, LCMS: 98%) as an off white solid. Note: Structure of desired isomer was confirmed by NOESY experiment. (LCMS m/z: 393.59 [M+H]+).


Step-4: A stirred solution of tert-butyl 2-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-4-yl)oxy)acetate (200 mg, 0.51 mmol) in DCM (5 mL) was treated with TFA (1.5 mL) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 3 h, monitored by TLC (Mobile phase: 40% EtOAc in Hexane, Rf:0.14, TLC detection: UV). All the volatiles were removed from reaction mixture and diluted with saturated solution of NaHCO3 (30 mL) and EtOAc (40 mL). The organic layer was separated and washed with saturated solution of NaHCO3 (2×30 mL). Finally, water phase was separated and organic phase (Observed precipitation) was directly evaporated under reduced pressure to get crude compound (800 mg). The crude compound was treated with 3 N solution of formic acid until to get semi-transparent solution and to attain pH 5. Then it was diluted with brine solution (10 mL) and extracted in EtOAc (2×25 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to get crude compound (144 mg, LCMS: 98%) as an off white solid. The crude compound was purified by prep HPLC Method H17. The pure fractions were concentrated under reduced pressure and traces of solvents were removed under high vacuum to afford 2-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-4-yl)oxy)acetic acid (Y: 80 mg, LCMS: 99%) as a white fluffy solid.


Example 20: Synthesis of 2-[[[1-[4-(trifluoromethyl)phenyl]indazol-3-yl]amino]methyl]prop-2-enoic acid (Cpd. No. 070)



embedded image


Step-1: In a glass screw-cap pressure vessel, a solution of 1H-indazol-3-amine (1.5 g, 12.26 mmol) in 1,4-dioxane (19 mL) was treated with 1-iodo-4-(trifluoromethyl)benzene (3.98 g, 14.64 mmol), CuI (429 mg, 2.25 mmol) and K2CO3 (3.11 g, 22.53 mmol) at room temperature. The reaction mixture was degassed by bubbling argon for 5 min, then added trans-N,N-dimethylcyclohexane-1,2-diamine (1.31 g, 9.01 mmol) at room temperature. The reaction mixture was heated to stir at 100° C. for 4 h, monitored by TLC (Mobile phase: 30% EtOAc in Hexane, Rf: 0.58, TLC detection: UV). The reaction mixture filtered on Celite bed and washed the bed with EtOAc (2×40 mL). Diluted the filtrate with water (60 mL), layers were separated and aqueous phase was extracted with EtOAc (30 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to get crude compound (1.8 g, LCMS: 84.7%) as dark brown liquid. The crude compound was triturated with n-pentane (2×15 ml) and dried under reduced pressure to get 1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-amine (1.5 g, LCMS: 95.2%) as brown solid. (LCMS m/z: 278.23 [M+H]+).


Step-2: A stirred solution of 1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-amine (600 mg, 2.16 mmol) in THF (10 mL) was treated with tert-butyl 2-(bromomethyl)acrylate (239.1 mg, 1.08 mmol) and N,N-diisopropylethylamine (307.2 mg, 2.38 mmol) at room temperature. The reaction mixture was heated to reflux for 16 h, monitored by TLC (Mobile phase: 10% EtOAc in Hexane, Rf: 0.32, TLC detection: UV). THF was evaporated under reduced pressure and reaction mixture was diluted with H2O (30 mL) and extracted in EtOAc (2×50 mL). The combined organic layer was dried over Na2SO4 and concentrated under reduced pressure to get crude product (700 mg, LCMS: 32% product+55% SM) as a brown semisolid. The crude compound was purified by Combi (100-200 mesh silica size) using 5-6% EtOAc in pet ether as eluent. Pure fractions were collected and concentrated under reduced pressure to get pure compound tert-butyl 2-(((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)amino)methyl)acrylate (250 mg, LCMS: 86%) as brown solid. (LCMS m/z: 362 [M-56]).


Step-3: A stirred solution of tert-butyl 2-(((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)amino)methyl)acrylate (250 mg, 0.6 mmol) in DCM (5 mL) was treated with TFA (2.5 mL) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h, monitored by TLC (Mobile phase: 50% EtOAc in Hexane, Rf: 0.18, TLC detection: UV). TFA was removed from reaction mixture under reduced pressure and the residue taken up in saturated solution of NaHCO3 (30 mL) and EtOAc (40 mL). The organic layer was separated, washed with saturated solution of NaHCO3 (30 mL) and dried over Na2SO4 and concentrated under reduced pressure to get crude compound (270 mg, LCMS: 96%) as a brown solid. The crude compound was purified by prep HPLC Method H17. The pure fractions were concentrated under reduced pressure and traces of solvents were removed under high vacuum to afford 2-((((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)amino)methyl)acrylic acid (Cpd. No. 070) (Y: 150 mg (60%), LCMS: 99%) as an off-white solid.


Example 21: Synthesis of N-[[1-(4,4-difluorocyclohexyl)indazol-3-yl]methyl]prop-2-enamide (Cpd. No. 077)



embedded image


Step-1: To a stirred solution of 4,4-difluorocyclohexan-1-ol (500 mg, 3.673 mmol) in DCM (10 mL) was added Et3N (0.774 mL, 5.509 mmol) followed by methanesulfonyl chloride (0.321 mL, 4.040 mmol) at 0° C. The reaction mixture was stirred for 3 h at RT under nitrogen atmosphere. The progress of the reaction was monitored by TLC (Mobile Phase: 10% EtOAc in Hexane, Rf=0.38, TLC detection: PMA Active). The reaction mixture was allowed to cool to 0° C., quenched with water (30 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine (2×30 mL), dried over Na2SO4 and concentrated under reduced pressure to afford 4,4-difluorocyclohexyl methanesulfonate (600 mg) as a yellow gummy oil. The product was used in next step without further purification.


Step-2: A stirred solution of 3-bromo-1H-indazole (600 mg, 3.045 mmol) in DMF (10 mL) was treated with cesium carbonate (1.984 g, 6.090 mmol) and 4,4-difluorocyclohexyl methanesulfonate (782.84 mg, 3.654 mmol) at 0° C. The reaction mixture was stirred at 100° C. for 16 h under nitrogen atmosphere. The progress of the reaction was monitored by TLC (Mobile Phase: 10% EtOAc in Hexane, Rf=0.65, TLC detection: UV Active). The reaction mixture was allowed to cool to RT and quenched with water (30 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine (2×30 mL), dried over Na2SO4 and concentrated under reduced pressure to afford the crude product (800 mg, LCMS: 34.9%) as pale brown gummy oil. The crude compound was purified by reverse phase column chromatography (C-18 column) and eluted with 50% ACN in H2O to afford 3-bromo (4,4-difluorocyclohexyl)-1H-indazole (680 mg, LCMS: 21.7% and 57.6% regioisomers) as a pale brown gummy oil. The product contains mixture of regioiosmers which were separated in the next step. (LCMS m/z: 315 [M+H]+).


Step-3: In a glass screw-cap pressure vessel, a solution of 3-bromo-1-(4,4-difluorocyclohexyl)-1H-indazole (550 mg, 1.745 mmol) in tert-butanol (5 mL) and water (5 mL) was treated with potassium (((tert-butoxycarbonyl)amino)methyl)trifluoroborate (517.149 mg, 2.181 mmol) and K2CO3 (1.445 g, 10.471 mmol) at room temperature. The reaction mixture was degassed by bubbling nitrogen for 10 min, added X-phosPd-G2 (274.335 mg, 0.349 mmol) and sealed with a teflon screw-cap. The reaction mixture was heated to 110° C. and stirred for 16 h, monitored by TLC (Mobile phase: 10% EtOAc in Hexane, Rf: 0.36, TLC detection: UV active). The reaction mixture was cooled to RT, filtered through a pad of Celite and the filter pad washed with EtOAc (2×50 mL). The filtrate was dried over Na2SO4 and concentrated under reduced pressure to afford crude product (800 mg, LCMS 36.7% & 7% of regioisomers) as pale brown gum. The crude product was purified by prep-H PLC Method H13, and the collected fractions concentrated and lyophilized to afford tert-butyl ((1-(4,4-difluorocyclohexyl)-1H-indazol-3-yl)methyl)carbamate (390 mg, LCMS: 97%) as a colourless semi solid. The structure was confirmed by NMR experiments. (LCMS m/z: 366 [M+H]+).


Step-4: A stirred solution of tert-butyl ((1-(4,4-difluorocyclohexyl)-1H-indazol-3-yl)methyl)carbamate (380 mg, 1.040 mmol) in DCM (10 mL) was treated with trifluoroacetic acid (1 mL) at 0° C. The resulting reaction mixture was allowed to warm to RT and stirred for 3 h under nitrogen atmosphere. The progress of the reaction was monitored by TLC (Mobile Phase: 5% MeOH in DCM, Rf=0.09, UV active). On completion, volatiles were evaporated and the residue dried under high vacuum to afford (1-(4,4-difluorocyclohexyl)-1H-indazol-3-yl)methanamine trifluoroacetate (500 mg, LCMS: 93.97%) as a pale brown gum. The product was used for the next step without any further purification. (LCMS m/z: 266 [M+H]+).


Step-5: A stirred solution of (1-(4,4-difluorocyclohexyl)-1H-indazol-3-yl)methanamine trifluoroacetate (470 mg, 1.772 mmol) in DCM (10 mL) was treated with Et3N (0.996 mL, 7.086 mmol) and acryloyl chloride (0.16 mL, 2.126 mmol) at 0° C. The reaction mixture was stirred at room temperature for 15 min under N2 atmosphere. The progress of the reaction was monitored by TLC (TLC Mobile Phase: 70% EtOAc in Hexane, Rf=0.85, TLC detection: UV Active) and LCMS. The reaction mixture was quenched with ice water (30 mL) and extracted with DCM (2×30 mL). The combined organic layer was washed with brine (2×30 mL), dried over Na2SO4 and concentrated under reduced pressure to afford crude product (350 mg, LCMS: 63.8%) as a pale brown gum. The crude product was purified by prep-HPLC Method H8 and collected fractions were concentrated and lyophilized to afford N-((1-(4,4-difluorocyclohexyl)-1H-indazol-3-yl)methyl)acrylamide (Cpd. No. 077) (122 mg, 21.6%, LCMS 98.7%) as a white solid.









TABLE 5







Analytical data for synthesized compounds of the disclosure











Cpd.
[M + H]+
LC/MS
RT

1H NMR



No.
(m/z) :
Method
(min.)
(δ ppm)














1
346
U2
1.15
(DMSO-d6) δ ppm: 8.82 (t, 1H), 8.02-8.08 (m, 2H),






7.92-8.01 (m, 4H), 7.57 (ddd, 1H), 7.30-7.37 (m, 1H),






6.29 (dd, 1H), 6.18 (dd, 1H), 5.65 (dd, 1H), 4.81 (d,






2H)


2
348
U2
1.15
(DMSO-d6) δ ppm: 8.48 (t, 1H), 8.01-8.09 (m, 2H),






7.90-8.00 (m, 4H), 7.57 (ddd, 1H), 7.33 (t, 1H), 4.71 (d,






2H), 2.15 (q, 2H), 1.03 (t, 3H)


3
370
U1
3.73
(DMSO-d6) δ ppm: 8.03-8.09 (m, 3H), 8.00 (d, 1H),






7.93-7.98 (m, 2H), 7.79 (br s, 1H), 7.60 (ddd, 1H),






7.34-7.41 (m, 1H), 4.63 (s, 2H), 2.98 (s, 3H)


4
362
U1
3.81
(DMSO-d6) δ ppm: 8.79 (t, 1H), 7.85-7.94 (m, 4H),






7.60 (d, 2H), 7.54 (ddd, 1H), 7.26-7.34 (m, 1H), 6.29






(dd, 1H), 6.18 (dd, 1H), 5.59-5.69 (m, 1H), 4.79 (d, 2H)


5
364
U1
3.84
(DMSO-d6) δ ppm: 8.46 (t, 1H), 7.85-7.95 (m, 4H),






7.60 (d, 2H), 7.53 (ddd, 1H), 7.25-7.34 (m, 1H), 4.69






(d, 2H), 2.15 (q, 2H), 1.03 (t, 3H)


6
386
U1
3.84
(DMSO-d6) δ ppm: 8.04 (d, 1H), 7.86-7.96 (m, 3H),






7.77 (br s, 1H), 7.60 (d, 2H), 7.56 (ddd, 1H), 7.30-7.38






(m, 1H), 4.61 (s, 2H), 2.97 (s, 3H)


7
396
L1
2.98
(DMSO-d6) δ ppm 8.04-8.06 (m, 2 H), 7.94-7.98 (m,






4 H), 7.56-7.60 (m, 1 H), 7.34-7.38 (m, 1 H), 4.53 (s,






2 H), 3.22-3.32 (m, 4 H), 2.18-2.26 (m, 2 H)


8
320
L1
2.85
(DMSO-d6) δ: 8.62-8.63 (m, 1 H), 8.31-8.33 (d, 1






H), 8.14-8.16 (d, 2 H), 7.99-8.01 (m, 3 H), 7.59-






7.63 (m, 1 H), 7.41-7.45 (t, 1 H), 2.86-2.87 (d, 3 H)


9
436
L1
2.83
(DMSO-d6) δ ppm: 8.14-8.16 (m, 2 H), 7.93-8.02






(m, 6 H), 7.63 (s, 1 H), 7.54-7.58 (t, 1 H), 7.31-7.35






(t, 1 H), 4.45-4.46 (d, 2 H), 3.76 (s, 3 H)


10
433
L1
2.51
(DMSO-d6) δ ppm: 8.77-8.4 (m, 3 H), 8.57-8.56 (m,






1H), 8.01-7.98 (m, 1 H), 7.95-7.90 (m, 5 H), 7.86-






7.84 (m, 1 H), 7.53-7.51 (m, 1 H), 7.53-7.49 (m, 1H),






7.33-7.33 (m, 2 H), 4.56 (s, 2H)


11
396
L1
2.59
(DMSO-d6) δ ppm: 8.07-8.05 (m, 3 H), 7.99-7.95






(m, 3 H), 7.87-7.84 (t, 1 H), 7.61-7.57 (m, 1 H), 7.35-






7.39 (t, 1 H), 4.64 (d, 2 H), 2.58-2.52 (m, 1 H), 0.94-






0.90 (m, 4 H)


12
384
L1
2.92
(DMSO-d6) δ: 12.35 (brs, 1 H), 8.25-8.29 (m, 3 H),






8.09-8.11 (d, 1 H), 8.02-8.04 (d, 2 H), 7.65-7.69






(m, 1 H), 7.50-7.54 (m, 1 H), 3.44 (s, 3 H)


13
399
L1
3.03
(DMSO-d6) δ ppm: 8.03-8.06 (m, 3 H), 7.92-8.00






(m, 4 H), 7.56-7.60 (m, 1 H), 7.34-7.38 (t, 1 H), 4.55-






4.56 (d, 2 H), 2.65 (s, 6 H)


14
410
L1
3.01
(DMSO-d6) δ: 12.35 (s, 1 H), 8.26-8.29 (m, 3 H),






8.08-8.11 (d, 1 H), 8.02-8.04 (d, 2 H), 7.65-7.69






(m, 1 H), 7.50-7.54 (t, 1 H), 3.16-3.20 (m, 1 H),






1.20-1.23 (m, 2H), 1.14-1.17 (m, 2 H)


15
380
L1
2.43
(DMSO-d6) δ: 8.38-8.41 (t, 1 H), 8.32-8.34 (d, 1






H), 8.14-8.16 (d, 2 H), 7.99-8.02 (m, 3 H), 7.61-






7.63 (m, 1 H), 7.42-7.46 (t, 1 H), 4.90-4.92 (d, 1 H),






4.62-4.65 (t, 1 H), 3.67-3.70 (m, 1 H), 3.49-3.56






(m, 1 H), 3.37-3.41 (m, 2 H), 3.26-3.29 (m, 1 H)


16
369
L1
2.39
(DMSO-d6) δ ppm: 8.09-8.11 (m, 1H), 8.03-8.05






(m, 2H), 7.92-7.98 (m, 3 H), 7.55-7.59 (t, 1 H), 7.32-






7.35 (t, 1 H), 7.09 (brs, 1 H), 4.52-4.57 (d, 2 H),






3.69 (brs, 1 H), 2.89 (brs, 3 H)


17
361
L1
2.79
(DMSO-d6) δ : 8.04-8.06 (d, 2 H), 7.93-7.99 (m, 4






H), 7.55-7.59 (m, 1 H), 7.31-7.34 (m, 1 H), 6.51 (s,






1 H), 4.68 (s, 2 H), 3.28-3.31 (m, 2 H), 3.19-3.23






(m, 2 H)


18
362
L1
2.95
(DMSO-d6) δ: 8.04-8.06 (d, 2 H), 7.91-8.00 (m, 4






H), 7.57-7.61 (m, 1 H), 7.34-7.38 (m, 1 H), 4.81 (s,






2 H), 4.25-4.29 (m, 2 H), 3.54-3.58 (m, 2 H)


19
347
L1
2.49
(DMSO-d6) δ ppm: 8.87-8.90 (t, 1 H), 8.73-8.74 (m,






1 H), 8.60-8.62 (d, 2 H), 8.40-8.42 (dd, 1 H), 7.94-






7.96 (d, 2 H), 7.43-7.46 (m, 1 H), 6.25-6.31 (m, 1 H),






6.15-6.19 (m, 1 H), 5.64-5.67 (m, 1 H), 4.79-4.80 (d,






2 H)


20
331
L2
3.07
(CDCl3) δ ppm: 10.5 (s, 1H), 8.24 (s, 1H), 8.00-8.01






(d, 1H), 7.91-7.93 (d, 2H), 7.82-7.84 (d, 2H), 7.69-7.71






(d, 1H), 7.29-7.33 (m, 1H), 7.22-7.26 (m, 1H), 6.62-






6.69 (m, 1H), 6.26-6.31 (dd, 1H), 5.76-5.79 (dd, 1H).


21
319
L3
4.98
(DMSO-d6) δ ppm: 10.11 (s, 1H), 8.10 (s, 1H), 7.98-






7.96 (d, 1H), 7.92-7.90 (d, 2H), 7.82-7.80 (d, 2H), 7.69-






7.67 (d, 1H), 7.30-7.26 (t, 1H), 7.23-7.19 (m, 1H), 2.15






(s, 3H).


22
263
L3
4.59
(DMSO-d6) δ ppm: 10.30 (s, 1H), 8.11 (s, 1H), 7.93-






7.95 (d, 1H), 7.57-7.64(m, 5H), 7.40-7.44 (m, 1H),






7.29-7.33 (m, 1H), 7.19-7.28 (m, 1H), 6.62-6.69 (m,






1H), 6.29-6.33 (dd, 1H), 5.80-5.83 (dd, 1H).


23
305
L2
3.23
(DMSO-d6) δ ppm: 10.28 (s, 1H), 8.10 (s, 1H), 7.94-






7.96 (d, 1H), 7.43-7.55 (m, 5H), 7.15-7.25 (m, 2H),






6.61-6.68 (m, 1H), 6.24-6.29 (m, 1H), 5.73-5.76 (m,






1H), 2.97-3.0 (t, 1H), 1.27 (s, 3H), 1.26 (s, 3H).


24
297
L2
3.04
(DMSO-d6) δ ppm: 10.31 (s, 1H), 8.13 (s, 1H), 7.98 (d,






1H), 7.62 (s, 4H), 7.55-7.58 (d, 1H), 7.25-7.29 (m, 1H),






7.18-7.22 (m, 1H), 6.61-6.68 (m, 1H), 6.25-6.29 (m,






1H), 5.74-5.78 (m, 1H).


25
297
L3
5.07
(DMSO-d6) δ ppm: 10.4 (s, 1H), 8.16 (s, 1H), 7.97-






7.99 (d, 1H), 7.57-7.66 (m, 4H), 7.43-7.46 (m, 1H),






7.19-7.30 (m, 2H), 6.62-6.68 (m, 1H), 6.25-6.30 (m,






1H), 5.75-5.78 (m, 1H).


26
339
L2
5.68
(DMSO-d6) δ ppm: 10.35 (s, 1H), 8.19 (s, 1H), 7.98-






8.00 (d, 1H), 7.93-7.95 (d, 2H), 7.74-7.76 (d, 2H), 7.63-






7.69 (m, 3H), 7.49-7.56 (m, 2H), 7.38-7.42 (t, 1H),






7.26-7.30 (t, 1H), 7.18-7.26 (t, 1H), 6.63-6.70 (q, 1H),






6.26-6.30 (dd, 1H), 5.75-5.78 (dd, 1H).


27
345
L2
2.94
(DMSO-d6) δ ppm: 8.46-8.49 (t, 1H), 7.92-7.94 (d, 2H),






7.81-7.83 (d, 2H), 7.66-7.72 (m, 3H), 7.24-7.29 (m,






1H), 7.17-7.21 (m, 1H), 6.1-6.21 (m, 2H), 5.58-5.61






(dd, 1H), 4.56-4.57 (d, 2H).


28
347
L2
2.93
(DMSO-d6) δ ppm: 8.12-8.15 (t, 1H), 7.92-7.94 (d, 2H),






7.80-7.82 (d, 2H), 7.66-7.7 (t, 3H), 7.24-7.28 (m, 1H),






7.16-7.20 (m, 1H), 4.46-4.47 (d, 2H), 2.09-2.10 (m,






2H), 1.00-1.04 (t, 3H).


29
345
L2
2.92
(DMSO-d6) δ ppm: 8.48-8.51 (t, 1H), 7.93-7.95 (d, 2H),






7.70-7.72 (d, 2H), 7.59-7.62 (m, 1H), 7.08-7.12 (m,






3H), 6.63 (s, 1H), 6.14-6.21 (m, 1H), 5.99-6.04 (dd,






1H), 5.54-5.57 (dd, 1H), 4.42-4.44 (d, 2H).


30
347
L2
2.93
(CDCl3) δ ppm: 7.79-7.85 (d, 2H), 7.60-7.64 (m, 1H),






7.51-7.53 (d, 2H), 7.11-7.19 (m, 3H), 6.62 (s, 1H), 5.49






(s, 1H), 4.53-4.54 (d, 2H), 2.08-2.14 (q, 2H), 1.05-1.08






(t, 3H).


31
359
L2
2.91
(CDCl3) δ ppm: 7.62-7.64 (d, 2H), 7.46-7.48 (d, 2H),






7.12-7.20 (m 3H), 7.00-7.02 (d, 1H), 6.56 (s, 1H), 6.05-






6.09 (dd, 1H), 5.69-5.76 (m, 1H), 5.44-5.51 (m, 4H),






4.66-4.68 (d, 2H).


32
359
L2
2.90
(CDCl3) δ ppm: 7.61-7.64 (m, 1H), 7.44-7.46 (d, 1H),






7.30-7.34 (m, 1H), 7.25-7.29 (m, 1H), 7.11-7.20 (m,






3H), 6.97-6.99 (d, 1H), 6.56 (s, 1H), 6.07-6.12 (dd,






1H), 5.69-5.76 (m, 1H), 5.49-5.52 (m, 2H), 5.44 (s,






2H), 4.69-4.70 (d, 2H).


33
347
L6
2.68
(DMSO-d6) δ ppm: 7.81-7.79 (d, 2H), 7.63-7.61 (d,






2H), 7.44-7.42 (d, 1H), 7.29-7.26 (m, 1H), 7.15-7.11 (t,






1H), 6.86-6.84 (d, 1H), 6.44-6.39 (m, 2H), 6.23-6.16






(m, 1H), 5.77-5.74 (d, 1H), 5.45-5.43 (d, 1H).


34
335
L2
2.54
(DMSO-d6) δ ppm: 8.91-8.89 (d, 1H), 7.96-7.94 (d,






2H), 7.68-7.66 (d, 2H), 7.33-7.23 (m, 2H), 7.12-7.08 (t,






1H), 6.85-6.83 (d, 1H), 5.28-5.26 (d, 1H), 1.91 (s, 3H).


35
333
U3
1.69
(DMSO-d6) δ ppm: 9.08 (br s, 1H), 8.36 (br s, 1H),






8.13 (s, 1H), 7.91 (d, 1H), 7.81 (d, 2H), 7.52-7.62 (m,






2H), 7.44-7.51 (m, 1H), 7.43 (d, 1H), 4.03 (s, 3H), 3.99






(br s, 2H).


36
410
U1
1.60
(DMSO-d6) δ ppm: 8.93 (s, 1H), 8.77 (d, 1H), 8.58 (d,






1H), 8.51 (dd, 1H), 8.49 (s, 1H), 8.43 (s, 1H), 7.88 (s,






2H), 7.78 (d, 2H), 7.70 (d, 1H), 7.51 (t, 2H), 7.34-7.45






(m, 2H), 5.52 (s, 2H), 3.69 (s, 2H).


37
346
U1
2.08
(DMSO-d6) δ ppm: 8.86 (d, 1H), 8.07 (d, 1H), 7.94 (d,






1H), 7.88 (s, 1H), 7.81 (d, 1H), 7.72 (d, 2H), 7.47-7.57






(m, 3H), 7.36-7.46 (m, 1H), 3.96 (s, 3H), 3.63 (s, 2H),






2.62 (d, 3H).


38
348
U1
1.30
(DMSO-d6) δ ppm: 8.50 (1H, s), 8.40 (1H, brs), 8.03






(1H, s), 7.89 (2H, s), 7.25-7.35 (2H, m), 7.23 (1H, s),






7.12 (2H, d), 6.89 (1H, t), 3.99 (3H, s), 3.91 (2H, s).


39
361
U1
2.10
(DMSO-d6) δ ppm: 8.32 (1H, d), 8.04 (1H, s), 7.91






(1H, d), 7.78 (2H, s), 7.72 (1H, d), 7.48 (1H, d), 7.23






(2H, t), 7.00 (2H, d), 6.80 (1H, t), 3.92 (3H, s), 3.57






(2H, s), 2.61 (3H, d).


40
347
L1
2.51
(DMSO-d6) δ ppm: 8.73-8.76 (t, 1H), 8.69-8.70 (dd,






1H), 8.46-8.49 (dd, 1H), 8.06-8.08 (d, 2H), 7.95-7.97






(d, 2H), 7.59-7.62 (m, 1H), 6.29-6.35 (dd, 1H), 6.11-






6.15 (dd, 1H), 5.60-5.63 (dd, 1H), 4.86-4.87 (d, 2H)


41
347
L1
1.86
(DMSO-d6) δ ppm: 9.262-9.264 (d, 1H), 8.93-8.96 (t,






1H), 8.54-8.55 (dd, 1H), 8.05-8.07 (d, 2H), 7.94-7.98






(m, 3H), 6.26-6.33 (m, 1H), 6.15-6.20 (m, 1H), 5.64-






5.67 (dd, 1H), 4.84-4.86 (d, 2H)


42
425
L2
2.89
(DMSO-d6) δ ppm: 8.09-7.92 (m, 5 H), 7.69 (d, 2 H),






7.36-7.34 (m, 1 H), 5.23 (s, 2 H), 4.59 (s, 2 H), 2.91






(s, 3H).


43
401
L2
2.86
(DMSO-d6) δ ppm: 8.81-8.78 (t, 1 H), 8.03-7.92 (m,






5 H), 7.61 (d, 1 H), 7.36-7.33 (m, 1 H), 6.32-6.25 (m,






1 H), 6.19-6.14 (m, 1 H), 5.65-5.62 (m, 1 H), 5.21 (s,






2 H), 4.77-4.76 (d, 2 H).


44
470
L5
3.42
(DMSO-d6) δ ppm: 8.02-8.00 (m, 2 H), 7.95-7.89






(m, 3 H), 7.50 (d, 1 H), 7.25-7.23 (m, 1 H), 4.58 (s, 2






H), 4.05-3.97 (m, 2 H), 3.94-3.79 (m, 2 H), 3.72-






3.66 (m, 1 H), 3.61-3.57 (m, 1 H), 2.95 (s, 3 H), 2.75






-2.71 (m, 1 H), 2.09-2.05 (m, 1 H), 1.73-1.69 (m, 1






H).


45
462
L3
5.73
(DMSO-d6) δ ppm: 8.06-8.01 (t, 3 H), 7.96-7.93 (m,






2 H), 7.76 (brs, 1 H), 7.708-7.03 (d, 1 H), 7.41-7.37






(m, 2 H), 7.34-7.32 (m, 1 H), 7.14-7.11 (t, 1 H),






7.03-7.00 (m, 2 H), 4.57 (s, 2 H), 2.93 (s, 3 H).


46
446
L5
3.42
(DMSO-d6) δ ppm: 8.01 (d, 2 H), 7.94-7.88 (m, 3 H),






7.41 (d, 1 H), 7.23-7.20 (m, 1 H), 6.33-6.26 (m, 1






H), 6.21-6.16 (m, 1 H), 5.69-5.65 (m, 1 H), 4.75 (s,






2 H), 4.02-3.91 (m, 2 H), 3.84-3.76 (m, 2 H), 3.71-






3.62 (m, 1 H), 3.57-3.55 (m, 1 H), 2.74-2.67 (m, 1






H), 2.08-2.03 (m, 1 H), 1.71-1.66 (m, 1 H).


47
446
L4
2.55
(DMSO-d6) δ ppm: 8.78 (t, 1 H), 8.02 (d, 2 H), 7.92-






7.88 (m, 3 H), 7.42 (d, 1 H), 7.22-7.19 (m, 1 H), 6.32-






6.22 (m, 1 H), 6.19-6.14 (m, 1 H), 5.65-5.62 (m, 1






H), 4.76 (d, 2 H), 4.02-3.91 (m, 2 H), 3.83-3.76 (m,






2 H), 3.70-3.64 (m, 1 H), 3.58-3.55 (m, 1 H), 2.72-






2.67 (m, 1 H), 2.10-2.01 (m, 1 H), 1.72-1.66 (m, 1






H).


48
446
L4
2.55
(DMSO-d6) δ ppm: 8.87 (t, 1 H), 8.01 (d, 2 H), 7.95(d,






2 H), 7.90 (d, 1 H), 7.40 (d, 1 H), 7.24-7.21 (m, 1 H),






6.33-6.26 (m, 1 H), 6.21-6.17 (m, 1 H), 5.70-5.67






(m, 1 H), 4.70 (d, 2 H), 4.02-3.91 (m, 2 H), 3.83-






3.76 (m, 2 H), 3.70-3.64 (m, 1 H), 3.58-3.55 (m, 1






H), 2.72-2.67 (m, 1 H), 2.08-2.03 (m, 1 H), 1.70-






1.66 (m, 1 H).


49
438
L3
5.72
(DMSO-d6) δ ppm: 8.04-7.94 (m, 5 H), 7.565-7.560






(d, 1 H), 7.41-7.37 (m, 2 H), 7.33-7.30 (m, 1 H),






7.15-7.14 (m, 1 H), 7.01-6.99 (m, 2 H), 6.26-6.21






(m, 1 H), 6.13-6.08 (m, 1 H), 5.65-5.62 (m, 1 H),






4.73 (s, 2 H).


50
402
L3
4.83
(DMSO-d6) δ ppm: 8.75-8.78 (t, 1 H), 8.00-8.02 (d,






2 H), 7.92-7.95 (d, 2 H), 7.78-7.80 (d, 1 H), 7.52 (s,






1 H), 6.98-7.01 (dd, 1 H), 6.23-6.30 (m, 1 H), 6.13-






6.18 (dd, 1 H), 5.62-5.65 (dd, 1 H), 4.72-4.73 (d, 2






H), 4.01-4.04 (m, 1 H), 0.81-0.86 (m, 2 H), 0.68-






0.71 (m, 2 H).


51
486
L3
3.75
(DMSO-d6) δ ppm: 11.35-10.65 (brs, 1 H), 8.82-






8.84 (t, 1 H), 8.04-8.10 (m, 3 H), 7.94-7.97 (d, 2 H),






7.86-7.87 (d, 1 H), 7.51-7.54 (dd, 1 H), 6.23-6.30






(dd, 1 H), 6.12-6.17 (dd, 1 H), 5.61-5.64 (dd, 1 H),






4.77-4.78 (d, 2 H), 4.34 (s, 1 H), 3.32 (s, 3 H).


52
466
L9
5.02
(DMSO-d6) δ ppm: 7.76-7.83 (q, 4 H), 7.65-7.67 (d, 1






H), 7.57-7.57 (d, 1 H), 7.47-7.52 (d, 1 H), 7.20-






7.22 (dd, 1 H), 6.46 (Broad s, 1 H), 5.26-5.33 (m, 3






H), 4.75-4.76 (d, 2 H), 2.92-3.00 (s, 3 H).


53
488
L6
1.97
(DMSO-d6) δ ppm: 11.18 (brs, 1 H), 8.46-8.49 (t, 1






H), 8.04-8.09 (m, 3 H), 7.94-7.96 (d, 2 H), 7.85-7.86






(d, 1 H), 7.51-7.54 (dd, 1 H), 4.67-4.69 (d, 2 H),






4.37 (s, 1 H), 3.30-3.40 (d, 3 H), 2.10-2.16 (q, 2 H),






0.98-1.01 (t, 3 H).


54
452
L6
2.41
(DMSO-d6) δ ppm: 8.80-8.77 (t, 1 H), 8.02-8.00 (d,






2 H), 7.92-7.90 (d, 3 H), 7.53-7.26 (m, 7 H), 6.32-






6.25 (m, 1 H), 6.20-6.15 (m, 1 H), 5.667-5.661 (m, 1






H), 5.15 (s, 2 H), 4.76-4.74 (d, 2 H).


55
454
L5
2.43
(DMSO-d6) δ ppm: 8.45 (s, 1 H), 8.02-7.90 (m, 5 H),






7.50 (s, 3 H), 7.41-7.26 (m, 4 H), 5.15 (s, 2 H), 4.66






(d, 2 H), 2.14 (d, 2 H), 1.03 (t, 3H).


56
476
L5
2.42
(DMSO-d6) δ ppm: 8.03 (d, 2 H), 7.93-7.91 (m, 3 H),






7.78 (s, 1 H), 7.63 (d, 1 H), 7.53-7.51 (m, 2 H), 7.43-






7.40 (m, 2 H), 7.37-7.28 (m, 2 H), 5.18 (s, 2 H),






4.58 (s, 2 H), 2.91 (s, 3 H).


57
395
L6
2.22
(DMSO-d6) δ ppm: 8.85 (s, 1H), 8.06-7.94 (m, 6 H),






7.61-7.57 (m, 1 H), 7.40-7.35 (t, 1 H), 4.86 (s, 2H),






4.73 (s, 2 H).


58
442
L9
5.09
(DMSO-d6) ppm: 12.85 (s, 1 H), 8.78-8.81 (t, 1 H),






8.00-8.02 (d, 2 H), 7.89-7.92 (m, 3 H), 7.66 (s, 1 H),






7.54-7.54 (d, 1 H), 7.23-7.26 (dd, 1 H), 6.40-6.40






(d, 1H), 6.26-6.33 (m, 1 H), 6.15-6.20 (dd, 1 H),






5.63-5.66 (dd, 1 H), 5.10 (s, 2 H), 4.75-4.77 (d, 2






H).


59
347
L2
1.85
(DMSO-d6) δ ppm: 9.45 (s, 1 H), 8.89-8.92 (t, 1 H),






8.44-8.46 (d, 1 H), 8.12-8.14 (d, 2 H), 7.96-7.98 (d,






2 H), 7.92-7.93 (m, 1 H), 6.24-6.31 (q, 1 H), 6.14-






6.19 (dd, 1 H), 5.63-5.66 (dd, 1 H), 4.83-4.81 (d, 2






H).


60
510
L9
4.71
(DMSO-d6) δ ppm: 11.11 (s, 1 H), 8.06-8.12 (m, 3 H),






7.95-7.97 (m, 3 H), 7.79 (Br s, 1 H), 7.54-7.57 (dd,






1 H), 4.62 (s, 2 H), 4.39 (s, 1 H), 3.41 (s, 3 H), 2.97 (s,






3 H).


61
426
L6
2.26
(DMSO-d6) δ ppm: 8.01-8.03 (dd, 2 H), 7.89-7.95






(m, 3 H), 7.75 (m, 1 H), 7.53-7.54 (d, 1 H), 7.01-7.04






(dd, 1 H), 4.54-4.55 (d, 2 H), 4.02-4.05 (m, 1 H),






2.95 (s, 3 H), 0.82-0.87 (m, 2 H), 0.69-0.73 (m, 2 H).


62
404
L6
2.31
(DMSO-d6) δ ppm: 8.43-8.45 (t, 1 H), 8.00-8.02 (dd,






2 H), 7.92-7.94 (dd, 2 H), 7.78-7.80 (dd, 1 H), 7.52-






7.525 (d, 1 H), 6.97-7.00 (dd, 1 H), 4.62-4.63 (d, 2






H), 4.00-4.05 (m, 1 H), 2.10-2.16 (q, 2 H), 1.00-






1.03 (t, 3 H), 0.82-0.86 (m, 2 H), 0.68-0.72 (m, 2 H).


63
446
L9
2.47
(DMSO-d6) δ ppm: 7.99-8.02 (d, 2 H), 7.90-7.93






(m, 3 H), 7.51-7.54 (m, 2 H), 7.24-7.27 (dd, 1 H),






4.84-4.85 (d, 2 H), 4.52-4.53 (d, 2 H), 3.57-3.58






(t, 1 H), 1.41 (s, 9 H).


64
402
L6
2.55
(DMSO-d6) δ ppm: 8.43-8.46 (t, 1 H), 8.00-8.02 (d,






2 H), 7.90-7.93 (dd, 3 H), 7.45-7.46 (d, 1 H), 7.24-






7.27 (dd, 1 H), 4.84-4.85 (d, 2 H), 4.65-4.66 (d, 2






H), 3.57-3.58 (t, 1 H), 2.13-2.19 (m, 2 H), 1.01-






1.05 (t, 3 H).


65
424
L6
2.55
(DMSO-d6) δ ppm: 8.02-8.04 (d, 2 H), 7.91-7.95






(m, 3 H), 7.77 (Broad s, 1 H), 7.57 (d, 1 H), 7.26-7.29






(dd, 1 H), 4.88-4.87 (d, 2 H), 4.58 (s, 2 H), 3.58-






3.59(t, 1 H), 2.94 (s, 3 H).


66
416
L6
2.71
(DMSO-d6) δ ppm: 8.76-8.79 (t, 1 H), 8.00-8.02 (d, 2






H), 7.88-7.92 (m, 3 H), 7.36-7.37 (d, 1 H), 7.19-






7.22 (dd, 1 H), 6.24-6.31 (m, 1 H), 6.13-6.18 (dd, 1






H), 5.62-5.65 (dd, 1 H), 4.73-4.75 (d, 2 H), 3.85-






3.87 (d, 2 H), 1.23-1.31 (m, 1 H), 0.57-0.61 (m, 2






H), 0.34-0.37 (m, 2 H).


67
418
L6
2.71
(DMSO-d6) δ ppm: 8.43-8.46 (t, 1 H), 7.99-8.01 (d,






2 H), 7.87-7.92 (m, 3 H), 7.35-7.36 (d, 1 H), 7.19-






7.22 (dd, 1 H), 4.63-4.65 (d, 2 H), 3.86-3.87 (d, 2






H), 2.11-2.17 (m, 2 H), 1.26-1.30 (m, 1 H), 1.00-






1.04 (t, 3 H), 0.61-0.62 (m, 2 H), 0.57-0.59 (m, 2






H).


68
440
L6
2.71
(DMSO-d6) δ ppm: 8.01-8.03 (d, 2 H), 7.89-7.92 (m,






3 H), 7.7 (brs s, 1 H), 7.47 (d, 1 H), 7.21-7.24 (dd, 1






H), 4.56 (s, 2 H), 3.89-3.91 (d, 2 H), 2.92 (s, 3 H),






1.26-1.32 (t, 1 H), 0.60-0.63 (m, 2 H), 0.58-0.60






(m, 2 H).


69
337
L6
2.44
(DMSO-d6): δ 8.44 (s, 1H), 8.04 (d, 2H), 7.94 (d, 2H),






7.54 (d, 1H), 7.45 (t, 1H), 6.69 (d, 1H), 4.89 (s, 2H).


70
362
L6
2.60
(DMSO-d6): δ 12.72 (brs, 1H), 7.99-7.97 (d, 1H), 7.93-






7.88 (m, 3H), 7.81-7.79 (d, 2H), 7.50 (t, 1H), 7.20 (t,






1H), 7.01 (brs, 1H), 6.14 (s, 1H), 5.82 (s, 1H), 4.20 (s,






2H).


71
414
L6
2.25
(DMSO-d6): δ 8.90 (t, 1H), 8.44 (s, 1H), 8.13 (d, 1H),






8.04 (m, 2H), 7.91 (m, 2H), 7.84 (d, 1H), 6.31-6.27 (m,






1H), 6.19-6.14 (m, 1H), 5.65 (dd, 1H), 4.84 (d, 2H).


72
402
L6
2.15
(DMSO-d6): δ 8.64 (t, 1H), 8.44 (s, 1H), 8.13 (d, 1H),






8.06-8.04 (m, 2H), 7.99-7.97 (m, 2H), 7.83 (d, 1H),






4.72 (d, 2H), 1.87 (s, 3H).


73
438
L6
2.22
(DMSO-d6): δ 8.53 (s, 1H), 8.14-8.16 (d, 1H), 8.05-






8.07 (d, 2H), 7.97-7.99 (d, 2H), 7.85-7.89 (m, 2H),






4.68-4.69 (d, 2H), 2.98 (s, 3H).


74
414
L3
5.98
(DMSO-d6): δ 8.87 (t, 1H), 8.23 (s, 1H), 8.19 (d, 1H),






8.07 (d, 2H), 7.98 (d, 2H), 7.65 (d, 1H), 6.30-6.24 (dd,






1H), 6.19-6.14 (dd, 1H), 5.66-5.63 (dd, 1H), 4.85-






4.83 (d, 2H).


75
402
L6
2.18
(DMSO-d6): δ 8.61 (t, 1H), 8.22 (s, 1H), 8.18 (d, 1H),






8.06 (d, 2H), 7.98 (d, 2H), 7.64 (d, 1H), 4.73 (d, 2H),






1.88 (s, 3H).


76
438
L6
2.26
(DMSO-d6): δ 8.28 (d, 1H), 8.25 (s, 1H), 8.08 (d, 2H),






7.99 (d, 2H), 7.85 (bs, 1H), 7.69-7.67 (dd, 1H), 4.67 (s,






2H), 2.99 (s, 3H).


77
320
L6
1.87
(DMSO-d6): δ 8.67 (t, 1H), 7.74-7.76 (d, 1H), 7.65-






7.67 (d, 1H), 7.37-7.41 (m, 1H), 7.13-7.10 (t, 1H), 6.21-






6.28 (dd, 1H), 6.12-6.17 (dd, 1H), 5.59-5.62 (dd, 1H),






4.83-4.86 (m, 1H), 4.66-4.68 (d, 2H), 2.09-2.21 (m,






6H), 1.98-2.01 (m, 2H).









Part B: Experimental Biology Procedures
Example 22: Activity of Compounds of the Disclosure in a Reporter Gene Assay for Measuring the Inhibition of YAP/TAZ-TEAD Transcription

Hek293T cells are cultured in DMEM supplemented with 10% fetal bovine serum, Sodium pyruvate, Sodium bicarbonate, L-glutamine. The cells are harvested and transiently transfected with TEAD-responsive element luciferase reporter. Transfected cells are plated in 384-wells plate containing pre-diluted compounds. After 24 hours incubation at 37° C./5% CO2, assay plates were cooled down to RT and levelled to an equal volume per well, prior to the addition of 25 uL luciferase substrate SteadyLite (Perkin Elmer)/well. The plate was shaken for 10 min at 600 rpm, centrifuged for 1 min at 500 rpm and measured with an Envision reader (PerkinElmer). The amount of relative light units produced by the TEAD reporter is used to calculated percent of inhibition.


The percent of reporter inhibition was calculated in the presence of a positive control inhibitor (100% inhibition) versus a condition with the presence of the vehicle basal activity of the reporter (0% inhibition). The ability of a test compound to inhibit this activity was determined as:





Percentage inhibition=[1−((RLU determined in the presence of vehicle−RLU determined for sample with test compound present) divided by (RLU determined in the presence of vehicle−RLU determined for sample with positive control inhibitor))]*100


The activities of example compounds tested are depicted in Table 6. The activity ranges A, B and C refer to IC50 values in the reporter gene assay as follows: “A”: IC50<1 μM; “B”: 1 μM≤IC50≤20 μM and “C”: IC50>20 μM; NT=not tested.









TABLE 6







Activities of compounds of the disclosure in the gene reporter


assay for measuring YAP/TAZ-TEAD transcription activity










Cpd. No.
EC50














1
A



2
B



3
A



4
A



5
B



6
B



7
B



8
C



9
C



10
C



11
B



12
B



13
B



14
C



15
B



16
B



17
A



18
A



19
A



20
A



21
A



22
B



23
A



24
A



25
B



26
A



27
A



28
B



29
A



30
C



31
A



32
A



33
B



34
C



35
C



36
C



37
C



38
C



39
C



40
A



41
A



42
C



43
A



44
B



45
B



46
A



47
A



48
A



49
A



50
A



51
A



52
B



53
C



54
A



55
C



56
B



57
B



58
A



59
A



60
C



61
B



62
C



63
A



64
C



65
C



66
A



67
C



68
A



69
C



70
A



71
A



72
B



73
B



74
A



75
C



76
B



77
A










Example 23: Activity of Compounds of the Disclosure in Mesothelioma Cell Line Proliferation Assays

Mesothelioma cell lines, NCI-H226 and NCI-H2052 (all sourced from the ATCC cell culture collection) are plated in 96-well plates (Corning® 96 Well White Polystyrene Microplate clear flat bottom, white polystyrene (TC-Treated)), at 1500 cells/well in full medium (RPMI 1640 ATCC modification with L-glutamine, HEPES, Phenol Red, Sodium Pyruvate, High glucose, Low sodium bicarbonate and 10% fetal bovine serum). Cells are incubated overnight at 37° C. in an incubator with 5% CO2. Then compounds, dissolved in DMSO, are added in dose-response. Cells are incubated with compound dilutions for another 6 days at 37° C. in an incubator with 5% CO2. Cell viability is quantitated using the ATPlite kit (Perkin-Elmer) and the luminescence is read-out using an Envision instrument (Perkin-Elmer). The amount of relative light units produced using the ATPlite kit is used to calculated percent of inhibition.


The activities of some example compounds are depicted in Table 7. The activity ranges A, B and C refer to EC50 values in the mesothelioma cell line proliferation assay as described as follows: “A”: EC50<1 μM; “B”: 1 μM≤EC50≤10 μM and “C”: EC50>10 μM; NT: not tested.









TABLE 7







Activities of a selection of compounds in the mesothelioma cell line proliferation assay









EC50









Cpd. No.
H226
H2052












1
A
A


3
C
C


4
A
A


19
A
A


20
B
A


21
A
B


27
C
B


29
A
A


31
A
B


40
A
A


43
A
A


46
A
A


47
A
A


48
A
A


49
A
A


50
A
A


54
A
A


58
A
A


59
A
A


63
A
A


66
A
A


71
A
A


74
A
A


77
A
A








Claims
  • 1. A compound of Formula I:
  • 2. A compound of Formula II:
  • 3. The compound of claim 1 or 2 of Formula III:
  • 4. The compound of any one of claims 1-3, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein: R2 is —(CH2)n—NR5aR5b; andR3 is selected from the group consisting of hydrogen and C1-C6 alkyl.
  • 5. The compound of any one of claims 1-4, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R5a is —C(═O)Z2 and R5b is hydrogen.
  • 6. The compound of any one of claims 1-5, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein: R2 is selected from the group consisting of hydrogen and C1-C6 alkyl; andR3 is —(CH2)p—NR6aR6b.
  • 7. The compound of any one of claims 1-6, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R6a is —C(═O)Z2 and R6b is hydrogen.
  • 8. The compound of claim 1 or 2 of Formula IV:
  • 9. The compound any one of claim 1, 2, or 8, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R3 is —(CH2)p—NR6aR6b.
  • 10. The compound of any one of claim 1, 2, 8, or 9, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein: R6a is selected from the group consisting of —C(═O)Z2 and —S(═O)2Z8; andR6b is hydrogen.
  • 11. The compound of claim 1 or 2 of Formula V:
  • 12. The compound of any one of claim 1, 2, or 11, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R3 is —(CH2)p—NR6aR6b.
  • 13. The compound of any one of claim 1, 2, 11, or 12, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein: R6a is selected from the group consisting of —C(═O)Z2; andR6b is hydrogen.
  • 14. The compound of any one of claim 1, 2, or 11-13, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein R1 is unsubstituted or substituted C6-C10 aryl, wherein one or more substituents are independently selected from the group consisting of halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, and —OZ1.
  • 15. The compound of any one of claim 1, 2, or 11-14, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, wherein: X is —CR7a═;X1 is —CR7b═;X2 is —CR7c═; andX3 is —CR7d═.
  • 16. The compound of claim 1, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of: N-[[5-(cyanomethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-[[rac-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-[[(3S)-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-[[(3R)-tetrahydrofuran-3-yl]methoxy]-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-phenoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[6-(cyclopropoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-(3-methyl-2,6-dioxo-pyrimidin-4-yl)oxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-benzyloxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-(1H-pyrazol-3-ylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[1-[4-(trifluoromethyl)phenyl]pyrazolo[3,4-c]pyridin-3-yl]methyl]prop-2-enamide;N-[[5-prop-2-ynoxy-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-(cyclopropylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[5-(cyclopropylmethoxy)-1-[4-(trifluoromethyl)phenyl]indazol yl]methyl]methanesulfonamide;2-[[[1-[4-(trifluoromethyl)phenyl]indazol-3-yl]amino]methyl]prop-2-enoic acid;N-[[5-(trifluoromethyl)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide;N-[[6-(trifluoromethyl)-1-[4-(trifluoromethyl)phenyl]indazol-3-yl]methyl]prop-2-enamide; andN-[[1-(4,4-difluorocyclohexyl)indazol-3-yl]methyl]prop-2-enamide.
  • 17. The compound of claim 2, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of: N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acrylamide;N-(1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)acetamide;N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)acrylamide;N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-3-yl)methyl)propionamide;N-(1-phenyl-1H-indol-3-yl)acrylamide;N-(1-(4-isopropylphenyl)-1H-indol-3-yl)acrylamide;N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)acrylamide;N-(1-(4-chlorophenyl)-1H-indol-3-yl)acrylamide;N-(1-(3-chlorophenyl)-1H-indol-3-yl)acrylamide;N-(1-([1,1′-biphenyl]-4-yl)-1H-indol-3-yl)acrylamide;N-((1-(4-(trifluoromethyl)phenyl)-1H-indol-2-yl)methyl)propionamide;N-((1-(3-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide;N-((1-(4-(trifluoromethyl)benzyl)-1H-indol-2-yl)methyl)acrylamide;N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)acrylamide;N-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)methanesulfonamide;N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acrylamide;N-(2-oxo-1-(4-(trifluoromethyl)phenyl)indolin-3-yl)acetamide; andN-((1-(4-(trifluoromethyl)phenyl)-1H-indazol-3-yl)methyl)propionamide.
  • 18. A compound of Formula VI:
  • 19. The compound of claim 18, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, selected from the group consisting of: 2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetic acid;N-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-phenylimidazo[1,2-a]pyridin-2-yl)acetamide;2-(6-phenyl-8-(1-(pyridin-3-ylmethyl)-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-2-yl)acetic acid;2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetic acid; andN-methyl-2-(8-(1-methyl-1H-pyrazol-3-yl)-6-(phenylamino)imidazo[1,2-a]pyridin-2-yl)acetamide.
  • 20. A pharmaceutical composition comprising the compound of any one of claims 1-19, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or solvate thereof, and a pharmaceutically acceptable carrier.
  • 21. The compound of any one of claims 1-19, or the pharmaceutical composition of claim 20, for use as a medicine.
  • 22. The compound of any one of claims 1-19, or the pharmaceutical composition of claim 20, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorder in an animal, mammal or human.
  • 23. The compound of any one of claims 1-19, or the pharmaceutical composition of claim 20, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorders is selected from the group comprising cancer, fibrosis and YAP/TAZ-TEAD activation mediated congenital disorders.
  • 24. The compound of any one of claims 1-19, or the pharmaceutical composition of claim 20, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorder selected from lung cancer, breast cancer, head and neck cancer, oesophageal cancer, kidney cancer, bladder cancer, colon cancer, ovarian cancer, cervical cancer, endometrial cancer, liver cancer (including but not limited to cholangiocarcinoma), skin cancer, pancreatic cancer, gastric cancer, brain cancer and prostate cancer, mesotheliomas, and/or sarcomas.
  • 25. The compound of any one of claims 1-19, or the pharmaceutical composition of claim 20, for use in the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorders selected from acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic, adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute T-cell leukemia, basal cell carcinoma, bile duct carcinoma, bronchogenic carcinoma, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer, endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, glioblastoma, gliosarcoma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, leukemia, liposarcoma, lymphagioendotheliosarcoma, lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, NUT midline carcinoma (NMC), non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenstrom's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor.
  • 26. A method for the prevention or treatment of a YAP/TAZ-TEAD activation mediated disorders in an animal, mammal or human comprising administering to said animal, mammal or human in need for such prevention or treatment an effective dose of the compounds of any one of claims 1-19.
  • 27. A method of treatment or prevention of YAP/TAZ-TEAD activation mediated disorder according to claim 26 to a patient in need thereof in combination with one or more other medicines selected from the group consisting of EGFR inhibitors, MEK inhibitors, AXL inhibitors, B-RAF inhibitors, and RAS inhibitors.
REFERENCE TO RELATED APPLICATIONS

The present application claims benefit of U.S. Provisional Application No. 63/293,537, filed Dec. 23, 2021, which is hereby incorporated by reference in its entirety.

Provisional Applications (1)
Number Date Country
63293537 Dec 2021 US