US20180325901A1 - Chiral diaryl macrocycles and uses thereof - Google Patents

Chiral diaryl macrocycles and uses thereof Download PDF

Info

Publication number
US20180325901A1
US20180325901A1 US15/745,915 US201615745915A US2018325901A1 US 20180325901 A1 US20180325901 A1 US 20180325901A1 US 201615745915 A US201615745915 A US 201615745915A US 2018325901 A1 US2018325901 A1 US 2018325901A1
Authority
US
United States
Prior art keywords
alkyl
alk
fusion protein
cancer
ros1
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/745,915
Other languages
English (en)
Inventor
Jingrong J. CUI
Yishan LI
Evan W. ROGERS
Dayong Zhai
Wei Deng
Zhongdong HUANG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Turning Point Therapeutics Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US15/745,915 priority Critical patent/US20180325901A1/en
Publication of US20180325901A1 publication Critical patent/US20180325901A1/en
Assigned to Turning Point Therapeutics, Inc. reassignment Turning Point Therapeutics, Inc. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: TP THERAPEUTICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/16Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/18Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Definitions

  • This disclosure relates to the use of certain diaryl macrocycle compounds, specifically (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1,15-ethenopyrazolo[4,3-f][1,4,8,10]benzoxatriazacyclotridecin-4(5H)-one in the treatment of disease in mammals.
  • This disclosure also relates to compositions including such compounds, and to methods of using such compositions in the treatment of diseases in mammals, especially in humans.
  • Protein kinases are key regulators for cell growth, proliferation and survival. Genetic and epigenetic alterations accumulate in cancer cells leading to abnormal activation of signal transduction pathways which drive malignant processes. (Manning, G.; Whyte, D. B.; Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase complement of the human genome. Science 2002, 298, 1912-1934). Pharmacological inhibition of these signaling pathways presents promising intervention opportunities for targeted cancer therapies. (Sawyers, C. Targeted cancer therapy. Nature 2004, 432, 294-297).
  • Anaplastic lymphoma kinase along with leukocyte tyrosine kinase (LTK), belongs to the insulin receptor (IR) superfamily of receptor tyrosine kinases.
  • ALK is mainly expressed in the central and peripheral nervous systems suggesting a potential role in normal development and function of the nervous system. (Pulford K, et al Cell Mol. Life Sci. 2004, 61, 2939).
  • ALK was first discovered as a fusion protein, NPM (nucleophosmin)-ALK encoded by a fusion gene arising from the t(2;5)(p23;q35) chromosomal translocation in anaplastic large cell lymphoma (ALCL) cell lines in 1994.
  • Mechanisms of drug resistance include target gene amplification or overexpression, development of secondary missense mutations, and use of alternative signaling pathway (so-called “bypass resistance”).
  • second-generation ALK inhibitors have been developed to be more potent against wild and many mutant ALKs.
  • One such mutation is the gatekeeper mutation ALK L1196M .
  • Ceritinib was approved by the US Food and Drug Administration for the treatment of patients with ALK-positive non-small cell lung cancer showing disease progression or who are intolerant to crizotinib. Although many second generation ALK inhibitors have been investigated in clinical trials, new ALK mutations resistant to the second generation ALK inhibitors have emerged.
  • the G1202R mutation has been found in tumors resistant to crizotinib, ceritinib, and alectinib.
  • crizotinib a tumor resistant to crizotinib, ceritinib, and alectinib.
  • Novel isoforms of ALK consisting primarily of the intracellular tyrosine kinase domain was found to express in ⁇ 11% of melanomas and sporadically in other human cancer types, but not in normal tissues (Wiesner T, et al Nature 2015, 526, 453-457). These new ALK isoforms stimulate multiple oncogenic signalling pathways, and are sensitive to ALK inhibitors, suggesting potential clinical benefits from ALK inhibition.
  • Non-small cell lung cancers harboring ALK gene rearrangements are sensitive to treatment with the ALK inhibitor crizotinib.
  • the mechanisms of resistance include ALK gene amplification, acquired ALK missense mutations, bypass pathway activation, and epithelial-mesenchymal transition (EMT) (Katayama R 2012).
  • EMT epithelial-mesenchymal transition
  • ALK rearranged NSCLCs are typically adenocarcinoma characterized by a solid signetring cell pattern that is frequently associated with a metastatic phenotype and linked to an epithelial-mesenchymal transition (EMT) phenotype.
  • EMT epithelial-mesenchymal transition
  • H2228 cell line with EML4-ALK v3 fusion gene displayed a mesenchymal phenotype with directly suppressing E-cadherin and up-regulating vimentin expression, as well as expression of other genes involved in EMT.
  • H2228 cell line confers intrinsic resistance to crizotinib and other ALK inhibitors.
  • ALK inhibition in patient-derived ALK models has been shown to up-regulate SRC activity.
  • the combination of a Src tyrosine kinase inhibitor with an ALK inhibitor was shown to effectively suppress downstream signaling, generated a synergistic inhibition effect, and re-sensitized the ALK inhibitors in the patient-derived ALK models in vitro and in vivo. (Crystal A S, Science.
  • ROS1 protein is a receptor tyrosine kinase, closely related to the ALK/LTK and insulin receptor kinase family. Although normal physiologic functions of human ROS1 kinase have not been fully understood, the abnormal expression and variable constitutively activating fusion forms of ROS1 kinase have been reported in a number of cancers including glioblastoma, non-small cell lung cancer, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma.
  • cancers including glioblastoma, non-small cell lung cancer, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendo
  • FIG-ROS1 fusion protein was the first fusion protein of ROS1 discovered in 2003 in a human glioblastoma multiforme.
  • FIG-ROS1 fusion protein was the first fusion protein of ROS1 discovered in 2003 in a human glioblastoma multiforme.
  • Several fusion proteins with ROS1 kinase including TPM3, SDC4, SLC34A2, CD74, EZR, and LRIG3 have been reported from human lung cancers, suggesting the oncogenic role of ROS1 kinase in lung cancers.
  • Takeuchi K et al Nat. Med.
  • ROS1 fusion partners including KDELR2, CCDC6, MSN, LIMA1, CLTC, NF ⁇ B2, NCOR2, CEP85L, TMEM106B, HLA-A, MYOSA, PPFIBP1, ERC1, PWWP2A, CLIP1, ZCCHC8, SHTN1, TFG, and YWHAE have been reported from various human cancers (Uquen A, et al Future Oncol. 2016, Jun. 3, Epub ahead of print) Taken together, ROS1 kinase is a promising molecular based target candidate for cancers with aberrant ROS kinase activities.
  • the ALK/MET/ROS1 inhibitor crizotinib has demonstrated marked efficacy in patients with NSCLC whose tumors are positive for ROS1 genetic abnormalities. (Shaw A T, et al N Engl J Med 2015, 372, 683). As expected ROS1 rearrangement-positive patients who responded to crizotinib eventually experienced disease progression. The secondary ROS1 G2032R mutation and bypass signaling are associated with the resistance. (Awad M M, et al N Engl J Med 2013, 368, 2395) It is desired to develop the next generation of ROS1 inhibitor to overcome the resistance.
  • Trks The tropomyosin-related receptor tyrosine kinases (Trks) are high-affinity receptors for neurotrophins (NTs), a nerve growth factor (NGF) family. Trk was originally cloned as an oncogene fused with the tropomyosin gene in the extracellular domain. The activating mutations caused by chromosomal rearrangements or mutations in TRK family have been reported in many cancers. (Vaishnavi A, et al Cancer Discov. 2015, 5, 25) Because Trks play important roles in pain sensation as well as tumour cell growth and survival signaling, inhibitors of Trk receptor kinases might provide benefit for pain and cancer treatment.
  • the Janus family of kinases include JAK1, JAK2, JAK3 and TYK2, and are cytoplastic non-receptor tyrosine kinases required for the physiologic signaling of cytokines and growth factors.
  • JAK2/Cardama A Aberrant regulation of JAK/STAT pathways has been implicated in multiple human pathological diseases, including cancer (JAK2) and rheumatoid arthritis (JAK1, JAK3).
  • a gain-of-function mutation of JAK2 has been discovered with high frequency in MPN patients.
  • JH2 pseudokinase domain of JAK2 leads to constitutively kinase activity.
  • Cells containing the JAK2V617F mutation acquire cytokine-independent growth ability and often become tumor, providing strong rationale for the development of JAK inhibitors as a targeted therapy.
  • hyperactivation of the JAK2/signal transducers and activators of transcription 3 is responsible for abnormal dendritic cell differentiation leading to abnormal dendritic cell differentiation and accumulation of immunosuppressive myeloid cells in cancer (Nefedova Y, et al. Cancer Res 2005, 65, 9525).
  • JAK2/STAT3 pathway In Pten-null senescent tumors, activation of the JAK2/STAT3 pathway establishes an immunosuppressive tumor microenvironment that contributes to tumor growth and chemoresistance (Toso A, et al. Cell Reports 2014, 9, 75). JAK2 gene fusions with the TEL(ETV6) (TEL-JAK2) and PCM1 genes have been found in leukemia patients. (Lacronique V, et al. Science 1997, 278, 5341, 1309-12. Reiter A, et al. Cancer Res.
  • JAK/STAT3 signaling pathway was aberrantly increased in EGFR inhibitor-resistant EGFR-mutant non-small cell lung cancer (NSCLC) cells, and JAK2 inhibition overcomes acquired resistance to EGFR inhibitors that support the use of combination therapy with JAK and EGFR inhibitors for the treatment of EGFR-dependent NSCLC.
  • NSCLC non-small cell lung cancer
  • c-Src is a nonreceptor tyrosine kinase.
  • the Src family (SFK) comprises of eight members in humans (Src, Fyn, Yes, Lyn, Lck, Hck, Blk and Fgr) with a molecular weight between 52-62 KDa.
  • Src and its family members are deregulated in many types of cancer.
  • Src is a key downstream transducer of many RTKs, including EGFR, HER2, and c-Met.
  • Src signaling has been implicated in conferring therapeutic resistance to targeted antiendocrine therapies, receptor tyrosine kinase therapies, traditional chemotherapies, and radiation therapies.
  • SRC can promote signaling from growth factor receptors in a number of ways including participation in signaling pathways required for DNA synthesis, control of receptor turn-over, actin cytoskeleton rearrangement, migration, adhesion, invasion, motility, and survival.
  • Resistance to EGFR inhibitors reportedly involves SRC activation and induction of epithelial-to-mesenchymal transition (EMT).
  • EMT epithelial-to-mesenchymal transition
  • the primary resistance to EGFR-TKIs is associated with higher levels of CRIPTO1 expression.
  • CRIPTO1 activated SRC and ZEB1 to promote EMT via microRNA-205 (miR-205) downregulation. Therefore, co-targeting EGFR and SRC may overcome intrinsic EGFR-inhibitor resistance in patients with CRIPTO1-positive, EGFR-mutated NSCLC. (Park, K-S, et al. J Clin Invest.
  • FAM Focal Adhesion Kinase
  • Src/FAK inhibitor may play important roles in combinatorial regimens in overcoming resistance to current anticancer therapies and in preventing metastatic recurrence, EMT and cancer treatment resistance.
  • AMP-activated protein kinase family member 5 also called NAUK1 is an upstream regulator of AMPK and limits protein synthesis via inhibition of rapamycin 1 (mTORC1) signalling pathway.
  • ARK5 maintains expression of mitochondrial respiratory chain complexes and respiratory capacity for efficient glutamine metabolism.
  • ARK5 is highly expressed in both primary NSCLC tissues and cell lines, that is functionally associated with NSCLC metastasis and a predictor of poor progriosi, for NSCLC patients.
  • ARK5 modulated the migration and invasion of NSCLC cells and played crucial roles in mTOR pathway.
  • L et al. Br J Cancer. 2014, 111(12):2316-27
  • ARK5 confers doxorubicinresistance in HCC via inducing EMT.
  • MYC MYC promotes cell growth and proliferation, and alters cellular metabolism.
  • Inhibition of ARK5 leads to a collapse of cellular ATP levels in cells expressing deregulated MYC, and prolongs survival in MYC-driven mouse models of hepatocellular carcinoma.
  • ARK5 inhibitor Targeting cellular energy homeostasis by ARK5 inhibitor is a valid therapeutic strategy to eliminate tumor cells with deregulated MYC expression.
  • Src is a non-receptor tyrosine kinase that is deregulated in many types of cancer, and a key downstream transducer of many RTKs, including EGFR, HER2, and c-Met. Activation of Src signaling has been implicated in conferring therapeutic resistance to targeted antiendocrine therapies, receptor tyrosine kinase therapies, traditional chemotherapies, and radiation therapies. (Zhang S, et al Trends Pharmacol Sci. 2012, 33, 122). Src inhibitor may play important roles in combinatorial regimens in overcoming resistance to current anticancer therapies and in preventing metastatic recurrence.
  • Cytoplasmic tyrosine kinases also known as non-receptor tyrosine kinases
  • Src family SFKs
  • SFKs Src family
  • Elevated SFK activity is found in more than 80% of human colorectal cancer (CRC) and this has been associated with poor clinical outcome.
  • CRC colorectal cancer
  • the SFK member Yes regulates specific oncogenic signalling pathways important for colon cancer progression that is not shared with c-Src. (Scancier F. et al. PLoS One.
  • WASF2FGR fusion genes were found in lung squamous carcinoma, ovarian serous cystadenocarcinoma, and skin cutaneous melanoma.
  • Estrogen receptorpositive (ER + ) breast cancers adapt to hormone deprivation and become resistant to antiestrogen therapy.
  • Mutations in the inhibitory SH2 domain of the SRC family kinase (SFK) LYN were related to ER + tumors that remained highly proliferative after treatment with the aromatase inhibitor letrozole. LYN was upregulated in multiple ER + breast cancer lines resistant to long-term estrogen deprivation.
  • the Src family kinase FYN is involved in signal transduction pathways in the nervous system, as well as the development and activation of T lymphocytes under normal physiological conditions. Activation of Fyn is observed in various cancers, including melanoma, glioblastoma, squamous cell carcinoma, prostate and breast cancers. (Elias D., et al. Pharmacological Research 2015, 100, 250-254) Fyn was upregulated in tamoxifen-resistant breast cancer cell lines and plays a key role in the resistance mechanism.
  • Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of aggressive non Hodgkin lymphomas with poor prognosis.
  • FYN activating mutations were found in PTCL, and promoted the growth of cells transformed via expression of activated FYN mutant alleles.
  • SRC kinase inhibitors may play important roles in the treatment of PTCLs. (Couronne L, et al. Blood 2013, 122, 811).
  • DDRs Discoidin domain receptors
  • matrix collagens have been implicated in numerous cellular functions such as proliferation, differentiation, adhesion, migration, and invasion.
  • DDRs play a role in cancer progression by regulating the interactions of tumor cells with their surrounding collagen matrix.
  • DDR1 is a direct p53 transcriptional target, and the activation of DDR1 is associated with p53-dependent DNA damage.
  • DDR1 activated the MAPK cascade in a Ras-dependent manner. Inhibition of DDR1 function led to increased apoptosis of wild-type p53-containing cells in response to genotoxic stress through a caspase-dependent pathway.
  • DDRs were identified as one of several major activated tyrosine kinases carrying somatic mutations in lung cancer (Hammerman P S, et al. Cancer Discov. 2011, 1, 78-89.), serous and clear cell endometrial cancer (Rudd M L, et al. BMC Cancer 2014, 14, 884), as well as in acute myeloid leukemia. (Tomasson M H, et al. Blood 2008, 111:4797-4808) Advanced Kirsten rat sarcoma viral oncogene homolog (KRAS)-mutant lung adenocarcinoma is challenging because of a lack of effective targeted therapies.
  • KRAS viral oncogene homolog
  • New compounds with polypharmacology profiles are also desired for targeting the primary oncogene drivers and their acquired resistance mechanisms including secondary mutations, bypath signaling, EMT, cancer stemness, and metastasis.
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are defined as described herein have been shown to have activity against wild-type and mutant ALK (anaplastic lymphoma kinase), wild-type and mutant ROS1 (ROS1 proto-oncogene receptor tyrosine kinase), the TRK family of kinases (tropomyosin-related receptor tyrosine kinases, TRKA/B/C), JAK2 of the Janus family of kinases and SRC (c-Src family of protein tyrosine kinases (SFKs)).
  • ALK anaplastic lymphoma kinase
  • ROS1 ROS1 proto-oncogene receptor tyrosine kinase
  • TRK family of kinases tropomyosin-related receptor tyrosine kinases
  • JAK2 JAK2 of the Janus family of kinases
  • SRC c
  • ALK anaplastic lymphoma kinase
  • ROS1 ROS1 proto-oncogene receptor tyrosine kinase
  • TRKA/B/C the TRK family of kinases
  • JAK2 JAK2 of the Janus family of kinases
  • SRC c-Src family of protein tyrosine kinases
  • Compound 1 has properties, including anti-tumor properties, which are pharmacologically mediated through inhibition of receptor and non-receptor tyrosine kinases.
  • Compound 1 is disclosed in International Patent Application No. PCT/US2015/012597, which is incorporated herein by reference in its entirety.
  • the present disclosure provide a method of treating disease in a patient comprising, administering to the patient a therapeutically effective amount of a compound of the formula I
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N(R 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , or Z 7 is independently N, NH, or C(R 10 ), wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 Z 5 , Z 6 or Z 7 is N or NH;
  • the present disclosure provides a method of treating cancer in a patient previously shown to express a genetically altered tyrosine or serine/throenine kinase comprising, administering to the patient a therapeutically effective amount of a compound of the formula I
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N(R 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or C(R 10 ), wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or CF 3 , and
  • Z 1 , Z 2 , or Z 7 is N or NH;
  • the present disclosure provides a method of treating cancer in a patient comprising;
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N(R 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or)C(R 10 ), wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • the present disclosure provides a method of identifying a patient for treatment with a compound of the formula I
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N( 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or C(R 10 ), wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N(R 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or C(R 10 ), wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or —CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N(R 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or)C(R 10 , wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —O—C 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or —CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N( 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or)C(R 10 , wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —O—C 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or —CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • the present disclosure provides use of a compound of the formula I
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N( 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or)C(R 10 , wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or —CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • the present disclosure provide a use a compound of the formula I
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N( 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or C(R 10 ), wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or —CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • the present disclosure provides the use of a compound of the formula I
  • M is CR 4a or N
  • X 1 and X 2 are independently S, S(O), S(O) 2 , O or N( 9 );
  • R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 alkyl)C(O)C 1 -C
  • each of R 2 and R 3 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 6 -C 10 aryl, —C(O)OR 7 or —C(O)NR 7 R 8 ; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and C 6 -C 10 aryl is independently optionally substituted by deuterium, halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —NHC(O)C 1 -C 6 alkyl, —N(C 1 -C 6 al
  • R 4 , R 4a and R 5 are each independently H, fluoro, chloro, bromo, C 1 -C 6 alkyl, —OH, —CN, —OC 1 -C 6 alkyl, —NHC 1 -C 6 alkyl, —N(C 1 -C 6 alkyl) 2 or —CF 3 ;
  • R 6 is H, C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl, wherein each hydrogen atom in C 1 -C 6 alkyl or 3- to 7-membered heterocycloalkyl is independently optionally substituted by halogen, —OH, —CN, —OC 1 -C 6 alkyl, —NH 2 , —NH(C 1 -C 6 alkyl), —N(C 1 -C 6 alkyl) 2 , —CO 2 H, —CO 2 C 1 -C 6 alkyl, —CONH 2 , —CONH(C 1 -C 6 alkyl), —CON(C 1 -C 6 alkyl) 2 , cycloalkyl, or monocyclic heterocycloalkyl;
  • each R 7 and R 8 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl or heteroaryl;
  • each R 9 is independently H, deuterium, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl; wherein each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or —OR 7 ;
  • each Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is independently N, NH, or)C(R 10 , wherein each R 10 is independently H, deuterium, halogen, C 1 -C 6 alkyl, —OC 1 -C 6 alkyl, —OH, —NH 2 , —NH(C 1 -C 6 alkyl), —NH(phenyl), —NH(heteroaryl), —CN, or —CF 3 , and
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 or Z 7 is N or NH;
  • a pharmaceutically acceptable salt thereof for treating cancer in a patient previously shown to express a genetically altered tyrosine or serine/threonine kinase, wherein the patient has been previously treated with a cancer therapeutic, and the cancer has developed resistance to the cancer therapeutic.
  • the compound is (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1,15-ethenopyrazolo[4,3-f][1,4,8,10]benzoxatriaza-cyclotridecin-4(5H)-one, or a pharmaceutically acceptable salt thereof.
  • the disease is mediated by a tyrosine or serine/threonine kinase selected from the group consisting of ALK, ROS1, TRKA, TRKB, TRKC, JAK2, SRC, FAK, ARK5, and combinations thereof.
  • the disease is mediated by a receptor tyrosine kinase.
  • the receptor tyrosine kinase is selected from the group consisting of ALK, ROS1, TRKA, TRKB and TRKC.
  • the receptor tyrosine kinase is selected from the group consisting of ALK, ROS1, TRKA, TRKB and TRKC.
  • the disease is mediated by a non-receptor kinase.
  • the non-receptor kinase is JAK2, FYN, LYN, YES, FGR, SRC, FAK or ARK5.
  • the non-receptor kinase is JAK2, SRC, FAK or ARK5.
  • the disease is mediated by a non-receptor tyrosine kinase.
  • the non-receptor tyrosine kinase is JAK2, SRC or FAK.
  • the disease is mediated by a non-receptor serine/threonine kinase.
  • the non-receptor serine/threonine kinase is ARK5.
  • the disease is mediated by a protein tyrosine kinase.
  • the protein tyrosine kinase is TXK.
  • the disease is mediated by a discoidin domain receptor.
  • the discoidin domain receptor is DDR1.
  • the disease is selected from the group consisting of cancer, psoriasis, rheumatoid arthritis, polycythemia vera, essential thrombocythemia, ulcerative colitis, and myeloid metaplasia with myelofibrosis and pain.
  • the disease or cancer is a cancer mediated by ALK. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered ALK. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by an ALK gene and a fragment of a protein which will form coiled-coil interaction to facilitate the protein dimerization or oligomerization.
  • the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a gene selected from the group consisting of NPM, EML4, TPR, TFG, ATIC, CLTC1, TPM4, MSN ALO17 and MYH9.
  • the fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by an EML4 gene.
  • the genetically altered ALK is an EML4-ALK fusion protein.
  • the EML4-ALK fusion protein is a wild-type protein.
  • the EML4-ALK fusion protein comprises at least one resistance mutation. In some embodiments, the EML4-ALK fusion protein comprises at least one mutation selected from the group consisting of L1196M, G1202R, D1203R, L1152P/R, F1174C/L/V, C1156Y, I1171N, G1123S, S1206Y, G1269S/A, and 1151T insertion.
  • the fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a NPM gene.
  • the genetically altered ALK is a NPM-ALK fusion protein.
  • the fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a TPR gene.
  • the genetically altered ALK is a TPR-ALK fusion protein.
  • the TPR-ALK fusion protein is a wild-type protein.
  • the TPR-ALK fusion protein comprises at least one resistance mutation.
  • the TPR-ALK fusion protein comprises a L1196M point mutation.
  • the disease or cancer is a cancer mediated by ALK. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered ALK. In some embodiments, the disease or cancer is a cancer mediated by ALK having one or more point mutations.
  • the disease or cancer is a cancer mediated by ALK having one or more point mutations selected from the group consisting of R1050H, F1174C/I/L/S/V, F1245C/I/L/V, R1275L/Q, T1151M, M1166R, 11170N, 11170S, 11171N, I1183T, L1196M, A1200V, L1204F, L1240V, D1270G, Y1278S, R1192P, G1128A, G1286R, and T13431.
  • the point mutation is a mutation at F1174.
  • the point mutation is a mutation of ALK at F1245.
  • the point mutation is a mutation of ALK at R1275.
  • the disease or cancer is a cancer mediated by ROS1. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered ROS1. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by an ROS1 gene and a fragment of a protein which will form coiled-coil interaction to facilitate the protein dimerization or oligomerization.
  • the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a ROS1 gene and a fragment of a protein encoded by a gene selected from the group consisting of FIG, TPM3, SDC4, SLC34A2, CD74, EZR, and LRIG3.
  • the fusion protein comprises a fragment of a protein encoded by an ROS1 gene and a fragment of a protein encoded by a CD74 gene.
  • the genetically altered ROS1 is a CD74-ROS1 fusion protein.
  • the CD74-ROS1 fusion protein is a wild-type protein.
  • the CD74-ROS1 fusion protein comprises at least one resistance mutation. In some embodiments, the CD74-ROS1 fusion protein comprises a G2032R point mutation. In some embodiments, the CD74-ROS1 fusion protein comprises a L2026M point mutation. In some embodiments, the CD74-ROS1 fusion protein comprises a D2033N point mutation. In some embodiments, the genetically altered ROS1 is a SDC4-ROS1 fusion protein. In some embodiments, the SDC4-ROS1 fusion protein is a wild-type protein. In some embodiments, the SDC4-ROS1 fusion protein comprises at least one resistance mutation.
  • the SDC4-ROS1 fusion protein comprises a G2032R point mutation.
  • the genetically altered ROS1 is a SLC34A2-ROS1 fusion protein.
  • the SLC34A2-ROS1 fusion protein is a wild-type protein.
  • the SLC34A2-ROS1 fusion protein comprises at least one resistance mutation.
  • the SLC34A2-ROS1 fusion protein comprises a G2032R point mutation.
  • the disease or cancer is a cancer mediated by TRKA. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered TRKA. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKA gene and a fragment of a protein which will form coiled-coil interaction to facilitate the protein dimerization or oligomerization. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKA gene and a fragment of a protein encoded by a TPM3 gene.
  • the genetically altered TRKA is a TPM3-TRKA fusion protein.
  • the TPM3-TRKA fusion protein is a wild-type protein.
  • the TPM3-TRKA fusion protein comprises at least one resistance mutation.
  • the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKA gene and a fragment of a protein encoded by a LMNA gene.
  • the genetically altered TRKA is a LMNA-TRKA fusion protein.
  • the LMNA-TRKA fusion protein is a wild-type protein.
  • the LMNA-TRKA fusion protein comprises at least one resistance mutation.
  • the LMNA-TRKA fusion protein is a wild-type protein.
  • the LMNA-TRKA fusion protein comprises at least one resistance mutation comprising a G595R point mutation.
  • the disease or cancer is a cancer mediated by TRKB. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered TRKB. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKB gene and a fragment of a protein which will form coiled-coil interaction to facilitate the protein dimerization or oligomerization. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKB gene and a fragment of a protein encoded by a QKI gene or TEL gene.
  • the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKB gene and a fragment of a protein encoded by a QKI gene. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKB gene and a fragment of a protein encoded by a TEL gene. In some embodiments, the genetically altered TRKB is a QKI-TRKB or TEL-TRKB fusion protein. In some embodiments, the genetically altered TRKB is a TEL-TRKB fusion protein. In some embodiments, the genetically altered TRKB is a QKI-TRKB fusion protein.
  • the QKI-TRKB or TEL-TRKB fusion protein is a wild-type protein. In some embodiments, the QKI-TRKB fusion protein is a wild-type protein. In some embodiments, the TEL-TRKB fusion protein is a wild-type protein. In some embodiments, the QKI-TRKB or TEL-TRKB fusion protein comprises at least one resistance mutation. In some embodiments, the QKI-TRKB fusion protein comprises at least one resistance mutation. In some embodiments, the TEL-TRKB fusion protein comprises at least one resistance mutation. In some embodiments, the TEL-TRKB fusion protein comprises a G639R point mutation.
  • the disease or cancer is a cancer mediated by TRKC. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered TRKC. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKC gene and a fragment of a protein which will form coiled-coil interaction to facilitate the protein dimerization or oligomerization. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a TRKC gene and a fragment of a protein encoded by an ETV6 gene. In some embodiments, the genetically altered TRKC is an ETV6-TRKC fusion protein.
  • the ETV6-TRKC fusion protein is a wild-type protein. In some embodiments, the ETV6-TRKC fusion protein comprises at least one resistance mutation. In some embodiments, the ETV6-TRKC fusion protein comprises a G623R point mutation.
  • the disease or cancer is a cancer mediated by JAK1, JAK2 or JAK3. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered JAK2. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a JAK2 gene and a fragment of a protein which will form coiled-coil interaction to facilitate the protein dimerization or oligomerization. In some embodiments, the disease or cancer is a cancer mediated by a fusion protein comprising a fragment of a protein encoded by a JAK2 gene and a fragment of a protein encoded by a TEL or PCM1 gene.
  • the genetically altered JAK2 is a TEL-JAK2 fusion protein. In some embodiments, the genetically altered JAK2 is a PCM1-JAK2 fusion protein. In some embodiments, the disease or cancer is a cancer mediated by point mutation(s) of JAK2. In some embodiments, the genetically altered JAK2 has the JAK2V617F mutation.
  • the disease is pain. In some embodiments, the disease is pain mediated by TRKA, TRKB or TRKC. In some embodiments, the pain is mediated by TRKA. In some embodiments, the pain is mediated by TRKB. In some embodiments, the pain is mediated by TRKC. In some embodiments, the disease is selected from the group consisting of psoriasis, rheumatoid arthritis, polycythemia vera, essential thrombocythemia, ulcerative colitis, and myeloid metaplasia with myelofibrosis. In some embodiments, the disease or cancer is a cancer exhibiting bypass resistance.
  • the disease or cancer is a cancer mediated by FGR. In some embodiments, the disease or cancer is a cancer mediated by a genetically altered FGR. In some embodiments, the fusion protein comprises a fragment of a protein encoded by a FGR gene and a fragment of a protein encoded by a WASF2 gene. In some embodiments, the genetically altered FGR is a WASF2-FGR fusion protein. In some embodiments, the WASF2-FGR fusion protein is a wild-type protein. In some embodiments, the WASF2-FGR fusion protein comprises at least one resistance mutation.
  • the cancer is selected from the group consisting of ALCL, NSCLC, neuroblastoma, inflammatory myofibroblastic tumor, adult renal cell carcinoma, pediatric renal cell carcinoma, breast cancer, colonic adenocarcinoma, glioblastoma, glioblastoma multiforme and anaplastic thyroid cancer.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, intrahepatic cholangiocarcinoma, colorectal cancer, thyroid papillary cancer, spitzoid neoplasms, sarcoma, astrocytoma, brain lower grade glioma, secretory breast carcinoma, mammary analogue carcinoma, breast cancer, acute myeloid leukemia, congenital mesoblastic nephroma, congenital fibrosarcomas, Ph-like acute lymphoblastic leukemia, colon adenocarcinoma, thyroid carcinoma, skin cutaneous melanoma, head and neck squamous cell carcinoma and pediatric glioma.
  • the cancer is selected from the group consisting of NSCLC, neuroblastoma, breast cancer, colon cancer and prostate cancer. In some embodiments, the cancer is NSCLC. In some embodiments, the cancer is neuroblastoma. In some embodiments, the cancer is colorectal cancer.
  • the patient has been previously treated with a cancer therapeutic. In some embodiments, the patient has been previously treated with a cancer therapeutic, and the cancer has developed resistance to the cancer therapeutic. In some embodiments, the resistance is a primary intrinsic resistance. In some embodiments, the resistance is an acquired resistance from mutation(s). In some embodiments, the resistance is a bypass resistance. In some embodiments, the resistance is an EMT-based resistance.
  • a method of treating disease in a patient comprising, administering to the patient a therapeutically effective amount of (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1,15-ethenopyrazolo[4,3-f][1,4,8,10]benzoxatriazacyclotridecin-4(5H)-one, or a pharmaceutically acceptable salt thereof.
  • non-receptor kinase is JAK2, FYN, LYN, YES, FGR, SRC, FAK or ARK5, including the non-receptor tyrosine kinase JAK2, FYN, LYN, YES, FGR, SRC or FAK, or the non-receptor serine/threonine kinase ARK5.
  • the fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by an EML4 gene.
  • EML4-ALK fusion protein comprises at least one mutation selected from the group consisting of L1196M, G1202R, D1203R, L1152P/R, F1174C/L/V, C1156Y, I1171N, G1123S, S1206Y, G1269S/A, and 1151T insertion.
  • the fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a NPM gene.
  • the fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a TPR gene.
  • any one of claim 1 to 4 or 32 wherein the disease is a cancer mediated by ALK having one or more point mutations selected from the group consisting of R1050H, F1174C/I/L/S/V, F1245C/I/L/V, R1275L/Q, T1151M, M1166R, 11170N, 11170S, I1171N, I1183T, L1196M, A1200V, L1204F, L1240V, D1270G, Y1278S, R1192P, G1128A, G1286R, and T13431.
  • ALK having one or more point mutations selected from the group consisting of R1050H, F1174C/I/L/S/V, F1245C/I/L/V, R1275L/Q, T1151M, M1166R, 11170N, 11170S, I1171N, I1183T, L1196M, A1200V, L1204F, L12
  • the fusion protein comprises a fragment of a protein encoded by an ROS1 gene and a fragment of a protein encoded by a CD74 gene.
  • CD74-ROS1 fusion protein comprises a G2032R, L2026M or D2033N point mutation.
  • any one of clauses 1 to 4, 7, 8 or 39 to 54 wherein the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma.
  • TPM3-TRKA or LMNA-TRKA fusion protein comprises at least one resistance mutation, including a LMNA-TRKA fusion protein comprising a G595R point mutation.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, intrahepatic cholangiocarcinoma, colorectal cancer, thyroid papillary cancer, spitzoid neoplasms, sarcoma, astrocytoma, brain lower grade glioma, secretory breast carcinoma, mammary analogue carcinoma, breast cancer, acute myeloid leukemia, congenital mesoblastic nephroma, congenital fibrosarcomas, Ph-like acute lymphoblastic leukemia, colon adenocarcinoma, thyroid carcinoma, skin cutaneous melanoma, head and neck squamous cell carcinoma and pediatric glioma.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, intrahepatic
  • a method of treating cancer in a patient previously shown to express a genetically altered tyrosine or serine/threonine kinase comprising, administering to the patient a therapeutically effective amount of (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro- 1,15-ethenopyrazolo[4,3-f][1,4,8,10]benzoxatriazacyclotridecin-4(5H)-one, or a pharmaceutically acceptable salt thereof.
  • the genetically altered ALK is a fusion protein comprising a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a gene selected from the group consisting of NPM, EML4, TPR, TFG, ATIC, CLTC1, TPM4, MSN ALO17 and MYH9.
  • fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by an EML4 gene.
  • the EML4-ALK fusion protein comprises at least one mutation selected from the group consisting of L1196M, G1202R, D1203R, L1152P/R, F1174C/L/V, C1156Y, I1171N, G1123S, S1206Y, G1269S/A, and 1151T insertion.
  • fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a NPM gene.
  • fusion protein comprises a fragment of a protein encoded by an ALK gene and a fragment of a protein encoded by a TPR gene.
  • ROS1 is a fusion protein comprising a fragment of a protein encoded by an ROS1 gene and a fragment of a protein encoded by a gene selected from the group consisting of FIG, TPM3, SDC4, SLC34A2, CD74, EZR, and LRIG3.
  • fusion protein comprises a fragment of a protein encoded by an ROS1 gene and a fragment of a protein encoded by a CD74 gene.
  • CD74-ROS1 fusion protein comprises a G2032R, L2026M or D2033N point mutation.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme, NSCLC, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma.
  • TRK is a fusion protein comprising a fragment of a protein encoded by a TRKA gene and a fragment of a protein encoded by a TPM3 gene or LMNA gene.
  • the cancer is selected from the group consisting of glioblastoma, glioblastoma multiforme,
  • JAK is fusion protein comprising a fragment of a protein encoded by a JAK2 gene and a fragment of a protein encoded by a TEL or PCM1 gene.
  • a method of treating cancer in a patient comprising;
  • step of identifying comprises subjecting a patient sample to a test selected from the group consisting of FISH, IHC, PCR and gene sequencing.
  • a method of identifying a patient for treatment with (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1,15-ethenopyrazolo[4,3-f][1,4,8,10]benzoxatriaza-cyclotridecin-4(5H)-one, or a pharmaceutically acceptable salt thereof, comprising diagnosing the patient with a cancer mediated by a genetically altered tyrosine or serine/threonine kinase.
  • diagnosing comprises subjecting a patient sample to a biological test or biological assay selected from the group consisting of FISH, IHC, PCR and gene sequencing.
  • FIG. 1 shows the effect of Compound 1 on apoptosis of Karpas-299 cells.
  • FIG. lA shows Karpas-299 cell apoptosis after incubation in various concentrations of Compound 1 for 48 hours;
  • FIG. 1B shows Karpas-299 cells were collected, lysed and analyzed by SDS-PAGE and immunoblotted with PARP and Actin antibodies.
  • FIG. 2 shows the effect of Compound 1 on wound healing in HCC78 and HT-1080 cells.
  • FIG. 3 shows gels demonstrating that Compound 1 down-regulates EGFR expression.
  • FIG. 3A shows results after 4 hour exposure to Compound 1 at 0 nM, 30 nM, 100 nM, 300 nM and 1000 nM;
  • FIG. 3A shows results after 24 hour exposure to Compound 1 at 0 nM, 30 nM, 100 nM, 300 nM and 1000 nM.
  • FIG. 4 shows gels demonstrating that Compound 1 down-regulates CD44 expression as compared to the same experiment with crizotinib which does not show down-regulation of CD44 expression.
  • FIG. 4A shows results after exposure to Compound 1 at 0 nM, 30 nM, 100 nM, 300 nM and 1000 nM;
  • FIG. 4A shows results after exposure to crizotinib at 0 nM, 30 nM, 100 nM, 300 nM and 1000 nM.
  • FIG. 5 shows the effect of Compound 1 on tumor growth in the Karpas-299 in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 50 mg/kg.
  • FIG. 6 shows the stability of animal body weight in the Karpas-299 in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 50 mg/kg.
  • FIG. 7 shows the effect of Compound 1 on tumor growth in the NIH3T3 EML4-ALK WT in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 50 mg/kg.
  • FIG. 8 shows the stability of animal body weight in the NIH3T3 EML4-ALK WT in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 50 mg/kg.
  • FIG. 9 shows the effect of Compound 1 on tumor growth in the NIH3T3 SDC4-ROS1 WT in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 50 mg/kg.
  • FIG. 10 shows the effect of Compound 1 on tumor growth in the KM12 in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 11 shows the stability of animal body weight in the KM12 in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 12 shows the inhibition of NPM-ALK phosphorylation by Compound 1 in the Karpas-299 in vivo model.
  • FIG. 13 shows the effect of Compound 1 on tumor growth in the KM12 in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 1 mg/kg, (•) 3 mg/kg, (1) 15 mg/kg.
  • FIG. 14 shows the stability of animal body weight in the KM12 in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 1 mg/kg, (•) 3 mg/kg, (1) 15 mg/kg.
  • FIG. 15 shows the effect of Compound 1 on tumor growth in the Ba/F3 EML4-ALK WT in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 16 shows the stability of animal body weight in the Ba/F3 EML4-ALK WT in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 17 shows the effect of Compound 1 on tumor growth in the Ba/F3 EML4-ALK G1202R in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 18 shows the stability of animal body weight in the Ba/F3 EML4-ALK G1202R in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 19 shows the effect of Compound 1 on tumor growth in the Ba/F3 CD74-ROS1 WT in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 20 shows the stability of animal body weight in the Ba/F3 CD74-ROS1 WT in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 21 shows the effect of Compound 1 on tumor growth in the Ba/F3 CD74-ROS1 G2032R in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 22 shows the stability of animal body weight in the Ba/F3 CD74-ROS1 G2032R in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 15 mg/kg, (•) 75 mg/kg.
  • FIG. 23 shows the effect of Compound 1 on tumor growth in the NIH3T3 LMNA-TRKA WT in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 3 mg/kg, (•) 15 mg/kg.
  • FIG. 24 shows the stability of animal body weight in the NIH3T3 LMNA-TRKA WT in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 3 mg/kg, (•) 15 mg/kg.
  • FIG. 25 shows the effect of Compound 1 on tumor growth in the NIH3T3 LMNA-TRKA G595R in vivo model at various doses, ( ⁇ ) control, ( ⁇ ) 3 mg/kg, (•) 15 mg/kg, (1) 60 mg/kg.
  • FIG. 26 shows the stability of animal body weight in the NIH3T3 LMNA-TRKA G595R in vivo model following administration of Compound 1 at various doses, ( ⁇ ) control, ( ⁇ ) 3 mg/kg, (•) 15 mg/kg, (1) 60 mg/kg.
  • alkyl includes a chain of carbon atoms, which is optionally branched and contains from 1 to 20 carbon atoms. It is to be further understood that in certain embodiments, alkyl may be advantageously of limited length, including C 1 -C 12 , C 1 -C 10 , C 1 -C 9 , C 1 -C 8 , C 1 -C 7 , C 1 -C 6 , and C 1 -C 4 , Illustratively, such particularly limited length alkyl groups, including C 1 -C 8 , C 1 -C 7 , C 1 -C 6 , and C 1 -C 4 , and the like may be referred to as “lower alkyl.” Illustrative alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl,
  • Alkyl may be substituted or unsubstituted.
  • Typical substituent groups include cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, oxo, ( ⁇ O), thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, C-carboxy, O-carboxy, nitro, and amino, or as described in the various embodiments provided herein.
  • alkyl may be combined with other groups, such as those provided above, to form a functionalized alkyl.
  • the combination of an “alkyl” group, as described herein, with a “carboxy” group may be referred to as a “carboxyalkyl” group.
  • Other non-limiting examples include hydroxyalkyl, aminoalkyl, and the like.
  • alkenyl includes a chain of carbon atoms, which is optionally branched, and contains from 2 to 20 carbon atoms, and also includes at least one carbon-carbon double bond (i.e. C ⁇ C). It will be understood that in certain embodiments, alkenyl may be advantageously of limited length, including C 2 -C 12 , C 2 -C 9 , C 2 -C 8 , C 2 -C 7 , C 2 -C 6 , and C 2 -C 4 .
  • alkenyl groups including C 2 -C 8 , C 2 -C 7 , C 2 -C 6 , and C 2 -C 4 may be referred to as lower alkenyl.
  • Alkenyl may be unsubstituted, or substituted as described for alkyl or as described in the various embodiments provided herein.
  • Illustrative alkenyl groups include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 1-, 2-, or 3-butenyl, and the like.
  • alkynyl includes a chain of carbon atoms, which is optionally branched, and contains from 2 to 20 carbon atoms, and also includes at least one carbon-carbon triple bond (i.e. CC). It will be understood that in certain embodiments, alkynyl may each be advantageously of limited length, including C 2 -C 12 , C 2 -C 9 , C 2 -C 8 , C 2 -C 7 , C 2 -C 6 , and C 2 -C 4 .
  • alkynyl groups including C 2 -C 8 , C 2 -C 7 , C 2 -C 6 , and C 2 -C 4 may be referred to as lower alkynyl.
  • Alkenyl may be unsubstituted, or substituted as described for alkyl or as described in the various embodiments provided herein.
  • Illustrative alkenyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, 1-, 2-, or 3-butynyl, and the like.
  • aryl refers to an all-carbon monocyclic or fused-ring polycyclic groups of 6 to 12 carbon atoms having a completely conjugated pi-electron system. It will be understood that in certain embodiments, aryl may be advantageously of limited size such as C 6 -C 10 aryl. Illustrative aryl groups include, but are not limited to, phenyl, naphthalenyl and anthracenyl. The aryl group may be unsubstituted, or substituted as described for alkyl or as described in the various embodiments provided herein.
  • cycloalkyl refers to a 3 to 15 member all-carbon monocyclic ring, including an all-carbon 5-member/6-member or 6-member/6-member fused bicyclic ring, or a multicyclic fused ring (a “fused” ring system means that each ring in the system shares an adjacent pair of carbon atoms with each other ring in the system) group, where one or more of the rings may contain one or more double bonds but the cycloalkyl does not contain a completely conjugated pi-electron system.
  • cycloalkyl may be advantageously of limited size such as C 3 -C 13 , C 3 -C 9 , C 3 -C 6 and C 4 -C 6 .
  • Cycloalkyl may be unsubstituted, or substituted as described for alkyl or as described in the various embodiments provided herein.
  • Illustrative cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cycloheptyl, adamantyl, norbornyl, norbornenyl, 9H-fluoren-9-yl, and the like.
  • Illustrative examples of cycloalkyl groups shown in graphical representations include the following entities, in the form of properly bonded moieties:
  • heterocycloalkyl refers to a monocyclic or fused ring group having in the ring(s) from 3 to 12 ring atoms, in which at least one ring atom is a heteroatom, such as nitrogen, oxygen or sulfur, the remaining ring atoms being carbon atoms.
  • Heterocycloalkyl may optionally contain 1, 2, 3 or 4 heteroatoms.
  • Heterocycloalkyl may also have one of more double bonds, including double bonds to nitrogen (e.g. C ⁇ N or N ⁇ N) but does not contain a completely conjugated pi-electron system.
  • heterocycloalkyl may be advantageously of limited size such as 3- to 7-membered heterocycloalkyl, 5- to 7-membered heterocycloalkyl, and the like.
  • Heterocycloalkyl may be unsubstituted, or substituted as described for alkyl or as described in the various embodiments provided herein.
  • heterocycloalkyl groups include, but are not limited to, oxiranyl, thianaryl, azetidinyl, oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, 1,4-dioxanyl, morpholinyl, 1,4-dithianyl, piperazinyl, oxepanyl, 3,4-dihydro-2H-pyranyl, 5,6-dihydro-2H-pyranyl, 2H-pyranyl, 1, 2, 3, 4-tetrahydropyridinyl, and the like.
  • Illustrative examples of heterocycloalkyl groups shown in graphical representations include the following entities, in the form of properly bonded moieties:
  • heteroaryl refers to a monocyclic or fused ring group of 5 to 12 ring atoms containing one, two, three or four ring heteroatoms selected from nitrogen, oxygen and sulfur, the remaining ring atoms being carbon atoms, and also having a completely conjugated pi-electron system. It will be understood that in certain embodiments, heteroaryl may be advantageously of limited size such as 3- to 7-membered heteroaryl, 5- to 7-membered heteroaryl, and the like. Heteroaryl may be unsubstituted, or substituted as described for alkyl or as described in the various embodiments provided herein.
  • heteroaryl groups include, but are not limited to, pyrrolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, pyridinyl, pyrimidinyl, quinolinyl, isoquinolinyl, purinyl, tetrazolyl, triazinyl, pyrazinyl, tetrazinyl, quinazolinyl, quinoxalinyl, thienyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, triazolyl, benzimidazolyl, benzoxazolyl, benzthiazolyl, benzisoxazolyl, benzisothiazolyl and carbazoloyl, and the like.
  • Illustrative examples of heteroaryl groups shown in graphical representations include the following entities, in the form of properly bonded
  • hydroxy or “hydroxyl” refers to an —OH group.
  • alkoxy refers to both an —O-(alkyl) or an —O-(unsubstituted cycloalkyl) group. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • aryloxy refers to an —O-aryl or an —O-heteroaryl group. Representative examples include, but are not limited to, phenoxy, pyridinyloxy, furanyloxy, thienyloxy, pyrimidinyloxy, pyrazinyloxy, and the like, and the like.
  • mercapto refers to an —SH group.
  • alkylthio refers to an —S-(alkyl) or an —S-(unsubstituted cycloalkyl) group. Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, cyclopropylthio, cyclobutylthio, cyclopentylthio, cyclohexylthio, and the like.
  • arylthio refers to an -S-aryl or an -S-heteroaryl group. Representative examples include, but are not limited to, phenylthio, pyridinylthio, furanylthio, thienylthio, pyrimidinylthio, and the like.
  • halo or halogen refers to fluorine, chlorine, bromine or iodine.
  • cyano refers to a —CN group.
  • oxo represents a carbonyl oxygen.
  • a cyclopentyl substituted with oxo is cyclopentanone.
  • substituted means that the specified group or moiety bears one or more substituents.
  • unsubstituted means that the specified group bears no substituents.
  • substitution is meant to occur at any valency-allowed position on the system.
  • substituted means that the specified group or moiety bears one, two, or three substituents.
  • substituted means that the specified group or moiety bears one or two substituents.
  • substituted means the specified group or moiety bears one substituent.
  • each hydrogen atom in C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl is independently optionally substituted by C 1 -C 6 alkyl” means that an alkyl may be but need not be present on any of the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 7-membered heterocycloalkyl, C 6 -C 10 aryl, or mono- or bicyclic heteroaryl by
  • independently means that the subsequently described event or circumstance is to be read on its own relative to other similar events or circumstances.
  • the use of “independently optionally” means that each instance of a hydrogen atom on the group may be substituted by another group, where the groups replacing each of the hydrogen atoms may be the same or different.
  • the use of “independently” means that each of the groups can be selected from the set of possibilities separate from any other group, and the groups selected in the circumstance may be the same or different.
  • the term “pharmaceutically acceptable salt” refers to those salts which counter ions which may be used in pharmaceuticals. See, generally, S.M. Berge, et al., “Pharmaceutical Salts,” J. Pharm. Sci., 1977, 66, 1-19.
  • Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of subjects without undue toxicity, irritation, or allergic response.
  • a compound described herein may possess a sufficiently acidic group, a sufficiently basic group, both types of functional groups, or more than one of each type, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Such salts include:
  • acid addition salts which can be obtained by reaction of the free base of the parent compound with inorganic acids such as hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, sulfuric acid, and perchloric acid and the like, or with organic acids such as acetic acid, oxalic acid, (D) or (L) malic acid, maleic acid, methane sulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, succinic acid or malonic acid and the like; or
  • a metal ion e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion
  • organic base such as ethanolamine, diethanolamine, triethanolamine, trimethamine, N-methylglucamine, and the like.
  • Pharmaceutically acceptable salts are well known to those skilled in the art, and any such pharmaceutically acceptable salt may be contemplated in connection with the embodiments described herein.
  • Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzo
  • a pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or tartaric acid, an inorganic acid, such as hydrochloric acid,
  • the disclosure also relates to pharmaceutically acceptable prodrugs of the compounds of Formula I, and treatment methods employing such pharmaceutically acceptable prodrugs.
  • prodrug means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug on being brought to physiological pH is converted to the compound of Formula I).
  • a “pharmaceutically acceptable prodrug” is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs”, ed. H. Bundgaard, Elsevier, 1985.
  • any formula depicted herein is intended to represent a compound of that structural formula as well as certain variations or forms.
  • a formula given herein is intended to include a racemic form, or one or more enantiomeric, diastereomeric, or geometric isomers, or a mixture thereof.
  • any formula given herein is intended to refer also to a hydrate, solvate, or polymorph of such a compound, or a mixture thereof.
  • the term “genetically altered” refers to a permanent alteration in the DNA sequence that makes up a gene that can result in a change in the protein sequence encoded by the gene.
  • a gene that is “genetically altered” as described herein can possess changes in DNA sequence, and/or protein sequence encoded by the DNA sequence, that range in size; for example, a single nucleotide (a.k.a. a single nucleotide polymorphism, SNP or point mutation), a multiple nucleotide polymorphism (MNPs), a large segment of a chromosome that includes multiple genes, such as a gene fusion, and the like.
  • gene fusions include, but are not limited to, those which are the result of a chromosomal inversion in which a portion of a chromosomal DNA encoding one or more genes rearranges to provide a fusion of two genes not ordinarily in communication in the DNA sequence, such as EML4-ALK (see for example, Soda, M. et al., Nature, 2007, 448, 561-567), those which are the result of a deletion in the DNA sequence (an “interstitial deletion”) in which part of a DNA sequence of a chromosome is deleted to provide a fusion of two genes not ordinarily in communication in the DNA sequence, such as TMPRSS2-ERG (see for example, Yu J.
  • a “genetically altered” gene, or the protein encoded by such gene can occur as hereditary mutations which can be inherited from a parent and are sometimes referred to as germline mutations, or a “genetically altered” gene, or the protein encoded by such gene, can occur as an acquired (or somatic) mutation that occurs at some point during a person's life.
  • a “genetically altered” gene can be described as a de novo (new) mutation, and can be either hereditary or somatic. It will be further understood that “genetically altered” can refer to a situation in which more than one of the changes in DNA sequence described herein can occur in a patient simultaneously, such as a SNP (or point mutation) and a translocation.
  • Such situations can arise from, but are not solely the result of, so-called “acquired resistance” in which a patient having been treated with a kinase inhibitor can develop a mutation in the DNA sequence that reduces the effectiveness of the treatment.
  • acquired resistance mutations include the point mutation L1196M, G1202R, L1152P, F1174C, C1156Y, 11171N, G1269S, and the 1151T insertion that occur in the EML4-ALK gene fusion.
  • intrinsic resistance refers to the pre-existing resistance of disease cells, especially cancer cells, to drug treatment, especially chemotherapy treatment. It will be appreciated that intrinsic resistance can result in resistance of the cells to a single drug, a small group of structurally related drugs, or a several drugs of differing chemical structure (so-called “multidrug resistance” or “MDR”). (Monti, E. 2007. Molecular Determinants of Intrinsic Multidrug Resistance in Cancer Cells and Tumors In B. Teicher (Ed.), Cancer Drug Resistance (pp. 241-260). Totowa, N.J.: Humana Press Inc.). It will be appreciated that intrinsic resistance can be the result of one or more host-related factors and/or the genetic make-up of the cells.
  • Such factors include but are not limited to immunomodulation; pharmacogenetic factors such as failure to achieve optimal serum drugs levels due to altered ADME or low tolerance to drug-induced side effects; restricted drug access to the tumor site; and microenvironmental cues.
  • genetic make-up factors include, but are not limited to altered expression of drug transporters; qualitative alterations of drug target(s); quantitative alterations of drug target(s); changes in intracellular drug handling/metabolism; changes in DNA repair activities, and alteration in apoptotic pathways. (Gottesman, M. M., Annu. Rev. Med., 2002, 53, 516-527).
  • the term “therapeutically effective amount” refers to that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a patient, which includes alleviation of the symptoms of the disease or disorder being treated. In one aspect, the therapeutically effective amount is that which may treat or alleviate the disease or symptoms.
  • the specific therapeutically-effective dose level for any particular patient will depend upon a variety of factors, including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, gender and diet of the patient: the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidentally with the specific compound employed; and like factors.
  • An exemplary dose is in the range of about from about 0.1 mg to 1 g daily, or about 1 mg to 50 mg daily, or about 50 to 250 mg daily, or about 250 mg to 1 g daily.
  • the total dosage may be given in single or divided dosage units (e.g., BID, TID, QID).
  • disease includes, but is not limited to, cancer, pain, psoriasis, rheumatoid arthritis, polycythemia vera, essential thrombocythemia, ulcerative colitis, and myeloid metaplasia with myelofibrosis.
  • cancer includes, but is not limited to, ALCL, NSCLC, neuroblastoma, inflammatory myofibroblastic tumor, adult renal cell carcinoma, pediatric renal cell carcinoma, breast cancer, ER + breast cancer, colonic adenocarcinoma, glioblastoma, glioblastoma multiforme, anaplastic thyroid cancer, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, epithelioid hemangioendothelioma, intrahepatic cholangiocarcinoma, thyroid papillary cancer, spitzoid neoplasms, sarcoma, astrocytoma, brain lower grade glioma, secretory breast carcinoma, mammary analogue carcinoma, acute myeloid leukemia, congenital mesoblastic nephroma, congen
  • the methods described herein relate to the treatment of disease comprising administering to a patient in need of treatment a therapeutically effective amount of a compound having activity against at least one tyrosine kinase selected from the group consisting of ALK, ROS1, TRK, JAK and SRC.
  • the compound has activity against at least one tyrosine or serine/threonine kinase selected from the group consisting of ALK, ROS1, TRKA, TRKB, TRKC, JAK2, SRC, FAK and ARK5.
  • the compound has activity against at least two tyrosine or serine/threonine kinases selected from the group consisting of ALK, ROS1, TRKA, TRKB, TRKC, JAK2, SRC, FAK and ARK5.
  • the at least one, or the at least two of the tyrosine or serine/threonine kinases are genetically altered.
  • the compound is of the formula I
  • the disease can be any of a number of diseases associated with the tyrosine kinases described herein against which the compounds of the formula I have activity.
  • the methods described herein can be used for the treatment of diseases such as cancer, pain, psoriasis, rheumatoid arthritis, polycythemia vera, essential thrombocythemia, ulcerative colitis, myeloid metaplasia with myelofibrosis, and the like.
  • diseases such as cancer, pain, psoriasis, rheumatoid arthritis, polycythemia vera, essential thrombocythemia, ulcerative colitis, myeloid metaplasia with myelofibrosis, and the like.
  • the disease can be any disease associated with the activity of a tyrosine kinase described herein.
  • the disease can be any disease associated with a genetically altered tyrosine or serine/threonine kinase selected from the group consisting of ALK, ROS1, TRKA, TRKB, TRKC, JAK2, SRC, FAK and ARK5. It will be further appreciated that the disease can be any disease associated with up-regulation of JAK2, SRC, FAK or ARK5.
  • the disease is a cancer mediated by or associated with a tyrosine kinase. In some embodiments, the disease is a cancer mediated by or associated with a serine/threonine kinase.
  • the disease is a cancer mediated by or associated with a genetically altered tyrosine kinase. In some embodiments, the disease is a cancer mediated by or associated with a genetically altered serine/threonine kinase. In some embodiments, the disease is a cancer mediated by or associated with up-regulation of JAK2, SRC, FAK or ARK5. In some embodiments, the disease is a cancer mediated by or associated with a genetically altered tyrosine kinase selected from the group consisting of ALK, ROS1, TRKA, TRKB, TRKC, JAK2, SRC and FAK. In some embodiments, the disease is a cancer mediated by or associated with a genetically altered serine/threonine kinase, such as ARK5.
  • the cancer can be any cancer mediated by or associated with a genetically altered tyrosine kinase selected from the group consisting of ALK, ROS1, TRKA, TRKB, TRKC and JAK2, or up-regulation of JAK2, SRC, FAK or ARK5, including but not limited to, ALCL, NSCLC, neuroblastoma, inflammatory myofibroblastic tumor, adult renal cell carcinoma, pediatric renal cell carcinoma, breast cancer, colonic adenocarcinoma, glioblastoma, glioblastoma multiforme, anaplastic thyroid cancer, cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal cancer, inflammatory myofibroblastic tumor, angiosarcoma, epithelioid hemangioendothelioma, intrahepatic cholangiocarcinoma, thyroid papillary cancer, spitzoid neoplasms, sarcoma,
  • the present disclosure provides methods of treating disease in a patient that has received a prior treatment with one or more therapeutic agents.
  • the patient has been previously treated with one or more chemotherapeutic agents.
  • the patent has been previously treated with one or more chemotherapeutic agents and developed an acquired resistance to the treatment.
  • the patent has been previously treated with one or more chemotherapeutic agents and developed bypass resistance to the treatment.
  • the patent has been previously treated with one or more chemotherapeutic agents and developed bypass resistance to the treatment regulated by SRC or JAK2.
  • chemotherapeutic agents which the patient may be been treated with prior to treatment with one or more of the compounds described herein include but are not limited to kinase inhibitors, adrenocorticoids and corticosteroids, alkylating agents, peptide and peptidomimetic signal transduction inhibitors, antiandrogens, antiestrogens, androgens, aclamycin and aclamycin derivatives, estrogens, antimetabolites, platinum compounds, amanitins, plant alkaloids, mitomycins, discodermolides, microtubule inhibitors, epothilones, inflammatory and proinflammatory agents, purine analogs, pyrimidine analogs, camptothecins and dolastatins.
  • the chemotherapeutic agent the patient received previous to treatment with one or more compounds described herein can be one or more of afatinib, axitinib, alectinib, bosutinib, brigatini, cabozantinib, ceritinib, crizotinib, dabrefenib, dasatinib, erlotinib, everolimus, gefitinib, ibrutinib, imatinib, lapatinib, lenvatinib, nilotinib, nintedanib, palbociclib, pazopanib, ponatinib, regorafenib, ruxolitinib, sirolimus, sorafenib, sunitinib, tofacitinib, temsirolimus, trametinib, vandetanib, vemurafenib, met
  • the methods described herein provide treatment of a patient previously treated with a kinase inhibitor selected from the group consisting of afatinib, alectinib, axitinib, bosutinib, brigatini, cabozantinib, ceritinib, crizotinib, dabrefenib, dasatinib, erlotinib, everolimus, gefitinib, ibrutinib, imatinib, lapatinib, lenvatinib, nilotinib, nintedanib, palbociclib, pazopanib, ponatinib, regorafenib, ruxolitinib, sirolimus, sorafenib, sunitinib, tofacitinib, temsirolimus, trametinib, vandetanib and vemurafenib
  • compositions comprising the compounds described herein may further comprise one or more pharmaceutically-acceptable excipients.
  • a pharmaceutically-acceptable excipient is a substance that is non-toxic and otherwise biologically suitable for administration to a subject. Such excipients facilitate administration of the compounds described herein and are compatible with the active ingredient. Examples of pharmaceutically-acceptable excipients include stabilizers, lubricants, surfactants, diluents, anti-oxidants, binders, coloring agents, bulking agents, emulsifiers, or taste-modifying agents.
  • pharmaceutical compositions according to the invention are sterile compositions. Pharmaceutical compositions may be prepared using compounding techniques known or that become available to those skilled in the art.
  • compositions are also contemplated by the invention, including compositions that are in accord with national and local regulations governing such compositions.
  • compositions and compounds described herein may be formulated as solutions, emulsions, suspensions, or dispersions in suitable pharmaceutical solvents or carriers, or as pills, tablets, lozenges, suppositories, sachets, dragees, granules, powders, powders for reconstitution, or capsules along with solid carriers according to conventional methods known in the art for preparation of various dosage forms.
  • Pharmaceutical compositions of the invention may be administered by a suitable route of delivery, such as oral, parenteral, rectal, nasal, topical, or ocular routes, or by inhalation.
  • the compositions are formulated for intravenous or oral administration.
  • the compounds the invention may be provided in a solid form, such as a tablet or capsule, or as a solution, emulsion, or suspension.
  • the compounds of the invention may be formulated to yield a dosage of, e.g., from about 0.1 mg to 1 g daily, or about 1 mg to 50 mg daily, or about 50 to 250 mg daily, or about 250 mg to 1 g daily.
  • Oral tablets may include the active ingredient(s) mixed with compatible pharmaceutically acceptable excipients such as diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservative agents.
  • Suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sugar, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, and the like.
  • Exemplary liquid oral excipients include ethanol, glycerol, water, and the like.
  • Starch, polyvinyl-pyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose, and alginic acid are exemplary disintegrating agents.
  • Binding agents may include starch and gelatin.
  • the lubricating agent if present, may be magnesium stearate, stearic acid, or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract, or may be coated with an enteric coating.
  • Capsules for oral administration include hard and soft gelatin capsules.
  • active ingredient(s) may be mixed with a solid, semi-solid, or liquid diluent.
  • Soft gelatin capsules may be prepared by mixing the active ingredient with water, an oil, such as peanut oil or olive oil, liquid paraffin, a mixture of mono and di-glycerides of short chain fatty acids, polyethylene glycol 400, or propylene glycol.
  • Liquids for oral administration may be in the form of suspensions, solutions, emulsions, or syrups, or may be lyophilized or presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid compositions may optionally contain: pharmaceutically-acceptable excipients such as suspending agents (for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminum stearate gel and the like); non-aqueous vehicles, e.g., oil (for example, almond oil or fractionated coconut oil), propylene glycol, ethyl alcohol, or water; preservatives (for example, methyl or propyl p-hydroxybenzoate or sorbic acid); wetting agents such as lecithin; and, if desired, flavoring or coloring agents.
  • suspending agents for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethyl
  • the agents of the invention may be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity or in parenterally acceptable oil.
  • Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride.
  • Such forms may be presented in unit-dose form such as ampoules or disposable injection devices, in multi- dose forms such as vials from which the appropriate dose may be withdrawn, or in a solid form or pre-concentrate that can be used to prepare an injectable formulation.
  • Illustrative infusion doses range from about 1 to 1000m/kg/minute of agent admixed with a pharmaceutical carrier over a period ranging from several minutes to several days.
  • inventive pharmaceutical compositions may be administered using, for example, a spray formulation also containing a suitable carrier.
  • inventive compositions may be formulated for rectal administration as a suppository.
  • the compounds of the present invention are preferably formulated as creams or ointments or a similar vehicle suitable for topical administration.
  • the inventive compounds may be mixed with a pharmaceutical carrier at a concentration of about 0.1% to about 10% of drug to vehicle.
  • Another mode of administering the agents of the invention may utilize a patch formulation to effect transdermal delivery.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, and 125 I, respectively.
  • Such isotopically labelled compounds are useful in metabolic studies (preferably with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques [such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • detection or imaging techniques such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements.
  • Isotopically labeled compounds of this disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • the compounds described herein may be used in pharmaceutical compositions or methods in combination with one or more additional active ingredients in the treatment of the diseases and disorders described herein.
  • Further additional active ingredients include other therapeutics or agents that mitigate adverse effects of therapies for the intended disease targets. Such combinations may serve to increase efficacy, ameliorate other disease symptoms, decrease one or more side effects, or decrease the required dose of an inventive compound.
  • the additional active ingredients may be administered in a separate pharmaceutical composition from a compound of the present invention or may be included with a compound of the present invention in a single pharmaceutical composition.
  • the additional active ingredients may be administered simultaneously with, prior to, or after administration of a compound of the present invention.
  • Combination agents include additional active ingredients are those that are known or discovered to be effective in treating the diseases and disorders described herein, including those active against another target associated with the disease.
  • compositions and formulations of the invention, as well as methods of treatment can further comprise other drugs or pharmaceuticals, e.g., other active agents useful for treating or palliative for the target diseases or related symptoms or conditions.
  • chemotherapeutic agents suitable for use in combination in the methods described herein include but are not limited to kinase inhibitors, adrenocorticoids and corticosteroids, alkylating agents, peptide and peptidomimetic signal transduction inhibitors, antiandrogens, antiestrogens, androgens, aclamycin and aclamycin derivatives, estrogens, antimetabolites, platinum compounds, amanitins, plant alkaloids, mitomycins, discodermolides, microtubule inhibitors, epothilones, inflammatory and proinflammatory agents, purine analogs, pyrimidine analogs, camptothecins and dolastatins.
  • chemotherapeutic agents suitable for combination treatments in the methods described herein include but are not limited to one or more of afatinib, alectinib, axitinib, bosutinib, brigatini, cabozantinib, ceritinib, crizotinib, dabrefenib, dasatinib, erlotinib, everolimus, gefitinib, ibrutinib, imatinib, lapatinib, lenvatinib, nilotinib, nintedanib, palbociclib, pazopanib, ponatinib, regorafenib, ruxolitinib, sirolimus, sorafenib, sunitinib, tofacitinib, temsirolimus, trametinib, vandetanib, vemurafenib, methot
  • chemotherapeutic agents suitable for combination treatments in the methods described herein include but are not limited to one or more kinase inhibitor selected from the group consisting of afatinib, alectinib, axitinib, bosutinib, brigatini, cabozantinib, ceritinib, crizotinib, dabrefenib, dasatinib, erlotinib, everolimus, gefitinib, ibrutinib, imatinib, lapatinib, lenvatinib, nilotinib, nintedanib, palbociclib, pazopanib, ponatinib, regorafenib, ruxolitinib, sirolimus, sorafenib, sunitinib, tofacitinib, temsirolimus, trametinib, vandetanib and vemur
  • the patient was previously treated with crizotinib.
  • suitable combination agents include anti-inflammatories such as NSAIDs.
  • the pharmaceutical compositions of the invention may additional comprise one or more of such active agents, and methods of treatment may additionally comprise administering an effective amount of one or more of such active agents.
  • the present disclosure provides methods for treating disease in a patient previously identified as having a genetically altered tyrosine or serine/threonine kinase, such as a gene fusion. In some embodiments, the present disclosure provides methods for treating cancer in a patient previously identified as having a genetically altered tyrosine or serine/threonine kinase, such as a gene fusion. In some embodiments, the present disclosure provides methods for treating disease in a patient comprising (i) identifying a genetically altered tyrosine or serine/threonine kinase in the patient, and (ii) administering to the patient a therapeutically effective amount of a compound useful in the treatment of such disease.
  • diagnosing or identifying a patient as having a genetically altered tyrosine or serine threonine kinase can be accomplished by any number of diagnostic tests known to one of skill in the art.
  • diagnostic tests include, but are not limited to, fluorescence in situ hybridization (FISH), polymerase chain reaction (PCR), immunohistochemistry (IHC), whole genome sequencing, next generation sequencing, and the like.
  • any of the methods known in the art and applicable to diagnosing a patient or identifying a patient in connection with the present disclosure involve the transformation of a biological sample from one state of matter to another by direct modification, synthesis or by direct non-covalent connection to provide a modified sample that can be used to determine whether the subject has or does not have a genetically altered tyrosine or serine threonine kinase.
  • “diagnosing” or “identifying” with respect to the disease state of a patient means applying a diagnostic test, such as FISH, PCR or IHC, to a biological sample obtained from the patient.
  • FISH is a test that “maps” the genetic material in a person's cells. This test can be used to visualize specific genes or portions of genes. FISH is a cytogenetic technique that uses fluorescent probes that bind to only those parts of the chromosome with a high degree of sequence complementarity. Such FISH tests can be used to identify a patient with a genetically altered tyrosine or serine/threonine kinase by any method known in the art, and such test can be used in combination with the methods described herein as either a means of prior identification of a patient for treatment, or the concomitant identification of a patient for treatment.
  • IHC refers to the process of detecting antigens (e.g., proteins) in cells of a tissue section by exploiting the principle of antibodies binding specifically to antigens in biological tissues.
  • Immunohistochemical staining is widely used in the diagnosis of abnormal cells such as those found in cancerous tumors.
  • Specific molecular markers are characteristic of particular cellular events such as proliferation or cell death (apoptosis).
  • Visualising an antibody-antigen interaction can be accomplished in a number of ways. In the most common instance, an antibody is conjugated to an enzyme, such as peroxidase, that can catalyse a colour-producing reaction. Alternatively, the antibody can also be tagged to a fluorophore, such as fluorescein or rhodamine.
  • IHC tests can be used to identify a patient with a genetically altered tyrosine or serine/threonine kinase by any method known in the art, and such test can be used in combination with the methods described herein as either a means of prior identification of a patient for treatment, or the concomitant identification of a patient for treatment.
  • PCR refers to a technology in molecular biology used to amplify a single copy or a few copies of a piece of DNA across several orders of magnitude, generating thousands to millions of copies of a particular DNA sequence.
  • Such PCR tests can be used to identify a patient with a genetically altered tyrosine or serine/threonine kinase by any method known in the art, and such test can be used in combination with the methods described herein as either a means of prior identification of a patient for treatment, or the concomitant identification of a patient for treatment.
  • whole genome sequencing or next-generation sequencing refers to a process that determines the complete DNA sequence of an organism's genome at a single time. This entails sequencing all of an organism's chromosomal DNA as well as DNA contained in the mitochondria.
  • whole genome sequencing tests can be used to identify a patient with a genetically altered tyrosine or serine/threonine kinase by any method known in the art, and such test can be used in combination with the methods described herein as either a means of prior identification of a patient for treatment, or the concomitant identification of a patient for treatment.
  • Step 2 Preparation of (R)—N—((R)-1-(5-fluoro-2-hydroxyphenyl)ethyl)-2-methylpropane-2-sulfinamide (2-3R)
  • Step 1 Preparation of ethyl (R)-5-((1-(5-fluoro-2-hydroxyphenyl)ethyl)amino)pyrazolo[1,5-a]pyrimidine-3-carboxylate (3)
  • reaction mixture was diluted with H 2 O (500 mL) at 16° C., and extracted with EtOAc (500 mLx3). The combined organic layers were washed with brine (500 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
  • Step 2 Preparation of ethyl 5-(((R)-1-(2-(((S)-1-((tert-butoxycarbonyl)amino)propan-2-yl)oxy)-5-fluorophenyl)ethyl)amino)pyrazolo[1,5-a]pyrimidine-3-carboxylate (4)
  • Step 3 Preparation of 5-(((R)-1-(2-(((S)-1-((tert-butoxycarbonyl)amino)propan-2-yl)oxy)-5-fluorophenyl)ethyl)amino)pyrazolo[1,5-a]pyrimidine-3-carboxylic acid (5)
  • Step 4 Preparation of 5-(((R)-1-(2-(((S)-1-aminopropan-2-yl)oxy)-5-fluorophenyl)ethyl)amino)pyrazolo[1,5-a]pyrimidine-3-carboxylic acid (6)
  • Step 5 Preparation of (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1,15-ethenopyrazolo[4,3-f][1,4,8,10]benzoxatriazacyclotridecin-4(5H)-one (Compound 1)
  • the reaction was stirred for 1 hour, and LC-MS showed the completion of the coupling reaction. The same process was repeated for 2 more times.
  • the final solution was stirred at ambient temperature for 63 hour (or until the reaction was shown to be completed by LC-MS).
  • the reaction was quenched by addition of aqueous Na 2 CO 3 solution (2M, 150 mL), and the mixture was stirred for 15 min, and extracted with DCM (3 ⁇ 150 mL).
  • kinase binding assays were performed at DiscoveRx using the general KINOMEscan K d Protocol (Fabian, M. A. et al., “A small molecule-kinase interaction map for clinical kinase inhibitors,” Nat. Biotechnol. 2005, 23(3):329-36).
  • kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32° C. until lysis. The lysates were centrifuged and filtered to remove cell debris.
  • the remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection.
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding.
  • Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in 1 ⁇ binding buffer (20% SeaBlock, 0.17 ⁇ PBS, 0.05% Tween 20, 6 mM DTT). All reactions were performed in polystyrene 96-well plates in a final volume of 0.135 mL. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1 ⁇ PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (1 ⁇ PBS, 0.05% Tween 20, 0.5 ⁇ M non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.
  • the biochemical kinase assay was performed at Reaction Biology Corporation (www.reactionbiology.com, Malvern, Pa.) following the procedures described in the reference (Anastassiadis T, et al Nat Biotechnol. 2011, 29, 1039). Specific kinase/substrate pairs along with required cofactors were prepared in reaction buffer; 20 mM Hepes pH 7.5, 10 mM MgCl2, 1 mM EGTA, 0.02% Brij35, 0.02 mg/ml BSA, 0.1 mM Na3VO4, 2 mM DTT, 1% DMSO (for specific details of individual kinase reaction components see Supplementary Table 2).
  • Human lung cancer cell line NCI-H2228 was obtained from ATCC.
  • Cell lines 293T, NIH3T3, Ba/F3, and HCC78 were purchased from DSMZ.
  • Karpas-299 cell line was purchased from Sigma.
  • KM12 cell line was obtained from NCI.
  • NIH3T3 was maintained in DMEM medium supplemented with 10% fetal bovine serum and 100 U/mL of penicillin/streptomycin.
  • HCC78, Karpas-299 and H2228 were maintained in RPMI-1640 supplemented with 10% fetal bovine serum with 100 U/mL of penicillin/streptomycin.
  • Ba/F3 cells were maintained in RPMI-1640 supplemented with 10% fetal bovine serum, 10% (Vol/Vol) conditioned media from the WIHI-3B myelomonocytic IL-3 secreting cells and 100 U/mL of penicillin/streptomycin.
  • BaF3 stable cell lines were maintained in RPMI-1640 supplemented with 10% fetal bovine serum, 100 U/mL of penicillin, and 0.5 ⁇ g/mL puromycin solution.
  • EML4-ALK gene (variant 1) was synthesized at GenScript and cloned into pCDH-CMV-MCS-EF1-Puro plasmid (System Biosciences, Inc). EML4-ALK point mutations G1202R, L1196M, L1152P, F1174C, C1156Y, 11171N, G1269S, and 1151T insertion were generated at GenScript by PCR and confirmed by sequencing. Ba/F3-EML4-ALK wild type and mutants were generated by transducing Ba/F3 cells with lentivirus containing EML4-ALK wide type or mutants. Stable cell lines were selected by puromycin treatment, followed by IL-3 withdrawal.
  • Ba/F3 cells were transduced with lentivirus supernatant in the presence of 8 ⁇ g/mL protamine sulfate.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin in the presence of IL3-containing medium RPMI1640, plus 10% FBS. After 10-12 days of selection, the surviving cells were further selected for IL3 independent growth.
  • SDC4-ROS1 wild type, CD74-ROS1 wild type, and their G2032R mutant genes were synthesized at GenScript and cloned into pCDH-CMV-MCS-EF1-Puro plasmid (System Biosciences, Inc), and confirmed by sequencing.
  • Ba/F3 SDC4-ROS1, CD74-ROS1 and corresponding G2032R mutants were generated by transducing Ba/F3 cells with lentivirus containing the fusion genes. Stable cell lines were selected by puromycin treatment, followed by IL-3 withdrawal. Briefly, 5 ⁇ 10 6 Ba/F3 cells were transduced with lentivirus supernatant in the presence of 8 ⁇ g/mL protamine sulfate.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin in the presense of IL3-containing medium RPMI1640, plus 10% FBS. After 10-12 days of selection, the surviving cells were further selected for IL3 independent growth.
  • Ba/F3 TPR-ALK and Ba/F3 TPR-ALK L1196M cell lines were created at Advanced Cellular Dynamics, Inc. and cell proliferation assays were performed there.
  • the ACD panel itself is comprised of 92 unique kinases individually expressed in a common lymphoid cell line (mouse Ba/F3 cells). Each cell line is dependent upon activity of the recombinant kinase for survival.
  • SDC4-ROS1 L2026M and D2033N mutant genes were synthesized at GenScript and cloned into pCDH-CMV-MCS-EF1-Puro plasmid (System Biosciences, Inc), and confirmed by sequencing.
  • Ba/F3 CD74-ROS1 L2026M and D2033N mutants were generated by transducing Ba/F3 cells with lentivirus containing the fusion genes. Stable cell lines were selected by puromycin treatment, followed by IL-3 withdrawal. Briefly, 5 ⁇ 10 6 Ba/F3 cells were transduced with lentivirus supernatant in the presence of 8 ⁇ g/mL protamine sulfate.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin in the presense of IL3-containing medium RPMI1640, plus 10% FBS. After 10-12 days of selection, the surviving cells were further selected for IL3 independent growth.
  • LMNA-TRKA wild type, and its G595R mutant genes were synthesized at GenScript and cloned into pCDH-CMV-MCS-EF1-Puro plasmid (System Biosciences, Inc), and confirmed by sequencing.
  • Ba/F3 LMNA-TRKA and corresponding G595R mutant were generated by transducing Ba/F3 cells with lentivirus containing the fusion genes. Stable cell lines were selected by puromycin treatment, followed by IL-3 withdrawal. Briefly, 5 ⁇ 10 6 Ba/F3 cells were transduced with lentivirus supernatant in the presence of 8 ⁇ g/mL protamine sulfate.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin in the presense of IL3-containing medium RPMI1640, plus 10% FBS. After 10-12 days of selection, the surviving cells were further selected for IL3 independent growth.
  • TEL-TRKB (also named as ETV6-TRKB) wild type and its G639R mutant genes were synthesized at GenScript and cloned into pCDH-CMV-MCS-EF1-Puro plasmid (System Biosciences, Inc), and confirmed by sequencing.
  • Ba/F3 TEL-TRKB and corresponding G639R mutants were generated by transducing Ba/F3 cells with lentivirus containing the fusion genes. Stable cell lines were selected by puromycin treatment, followed by IL-3 withdrawal. Briefly, 5 ⁇ 10 6 Ba/F3 cells were transduced with lentivirus supernatant in the presence of 8 ⁇ g/mL protamine sulfate.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin in the presense of IL3-containing medium RPMI1640, plus 10% FBS. After 10-12 days of selection, the surviving cells were further selected for IL3 independent growth.
  • TEL-TRKC also named as ETV6-TRKC
  • G623R mutant genes were synthesized at GenScript and cloned into pCDH-CMV-MCS-EF1-Puro plasmid (System Biosciences, Inc), and confirmed by sequencing.
  • Ba/F3 TEL-TRKC and corresponding G623R mutant were generated by transducing Ba/F3 cells with lentivirus containing the fusion genes. Stable cell lines were selected by puromycin treatment, followed by IL-3 withdrawal. Briefly, 5 ⁇ 10 6 Ba/F3 cells were transduced with lentivirus supernatant in the presence of 8 ⁇ g/mL protamine sulfate.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin in the presense of IL3-containing medium RPMI1640, plus 10% FBS. After 10-12 days of selection, the surviving cells were further selected for IL3 independent growth.
  • NIH3T3 ALK or ROS1 stable cell lines were generated by transducing the cells with lentivirus containing EML4-ALK wild type and G1202R mutant genes, SDC4-ROS1 wild type, CD74-ROS1 wild type and their G2032R mutant genes. The transduced cells were subsequently selected with 1 ⁇ g/mL puromycin.
  • NIH3T3 ROS1, or TRKA , or TRKB, or TRKC stable cell lines were generated by transducing the cells with lentivirus containing CD74-ROS1 L2026M and D2033N mutant genes, LMNA-TRKA wild type and G595R mutant genes, TEL-TRKB wild type and G639R mutant genes, TEL-TRKB wild type and G623R mutant genes, respectively.
  • the transduced cells were subsequently selected with 1 ⁇ g/mL puromycin.
  • the cell proliferation assay of Ba/F3 TPR-ALK and B a/F3 TPR-ALK L1196M cell lines were performed at Advanced Cellular Dynamics, Inc.
  • the inhibition of kinase activity leads to cell death, which is monitored via ATP concentration using CellTiter-Glo (Promega).
  • Cell lines were maintained in RPMI-1640 culture media containing 10% fetal calf serum and antibiotics. Cells in logarithmic-phase growth were harvested and 5,000 cells were distributed into each well of a 384-well plate in 50 ⁇ L of growth media. Parental cells (only) were seeded in the presence of 10 ng/mL IL3 to support cell growth and survival.
  • NSCLC cell line H2228 (harboring endogenous EML4-ALK fusion gene), HCC78 cells (harboring endogenous SLC34A2-ROS1 fusion gene), Karpas-299 cells (harboring endogenous NPM-ALK fusion gene) or SET-2 (harboring endogenous JAK2V617F activating mutation) were cultured in RPMI medium, and KM12 (harboring endogenous TPM3-TRKA fusion gene) cell line was cultured in DMEM medium, both supplemented with 10% fetal bovine serum and 100 U/mL of penicillin/streptomycin.
  • Ba/F3 and NIH3T3 cells stably expressing ALK or ROS1 (WT or mutant) were culture as mentioned above.
  • Half a million cells per well were seeded in 24 well plate for 24 hrs, and then treated with compounds for 4 hours.
  • Cells were collected after treatment and lysed in RIPA buffer (50 mM Tris, pH 7.4, 150 mM NaCl, 1% NP-40, 0.5% Deoxycholate, 0.1% SDS) supplemented with 10 mM EDTA, 1 ⁇ Halt protease and phosphatase inhibitors (Thermo Scientific).
  • Protein lysates (approximately 20 ⁇ g) was resolved on 4-12% Bolt Bis-Tris precasted gels with MES running buffer (Life Technologies), transferred to nitrocellulose membranes using Trans-Blot Turbo Transfer System (Bio-Rad) and detected with antibodies targeting phosphorylated ALK Y1604 (Cell Signaling Technology), total ALK (Cell Signaling Technology), phosphorylated ROS1 and total ROS1 (Cell Signaling Technology), phosphorylated TRK A/B (Cell Signaling Technology), total TRKA antibody (Santa Cruz Biotechnogy), phosphorylated STAT3 and STATS, total STAT3 and STATS (Cell Signaling Technology), phosphorylated AKT (Cell Signaling Technology), total AKT (Cell Signaling Technology), phosphorylated ERK (Cell Signaling Technology), total ERK (Cell Signaling Technology) and Tubulin (Sigma).
  • Antibodies were typically incubated overnight at 4° C. with gentle shake, followed by washes and incubation with the appropriate HRP-conjugated secondary antibodies.
  • Membranes were incubated with chemiluminescent substrate for 5 min at room temperature (SuperSignal West Femto, Thermo Scientific).
  • the chemiluminescent images were acquired with a C-DiGit Imaging System (LI-COR Biosciences).
  • the relative density of the chemiluminescent bands were quantified via Image Studio Digits from LICOR.
  • the half inhibitory concentration (IC 50 ) value is calculated using non-linear regression analysis through GraphPad Prism software (GraphPad, Inc., San Diego, Calif.).
  • NSCLC cell line H2228 (harboring endogenous EML4-ALK fusion gene) was cultured in RPMI medium supplemented with 10% fetal bovine serum and 100 U/mL of penicillin/streptomycin.
  • Half a million cells per well were seeded in 24 well plate for 24 hrs, and then treated with compounds for 4 hours.
  • Protein lysates (approximately 20 ⁇ g) was resolved on 4-12% Bolt Bis-Tris precasted gels with MES running buffer (Life Technologies), transferred to nitrocellulose membranes using Trans-Blot Turbo Transfer System (Bio-Rad) and detected with antibodies targeting phosphorylated phosphorylated ROS1 and total ROS1 (Cell Signaling Technology), phosphorylated TRK A/B (Cell Signaling Technology), total TRKA antibody (Santa Cruz Biotechnogy), phosphorylated SRC Y416 (Cell Signaling Technology), total SRC (Cell Signaling Technology), phosphorylated FAK Y576/577 (Cell Signaling Technology), total FAK (Cell Signaling Technology), phosphorylated paxillin Y118 (Cell Signaling Technology), total paxillin (Cell Signaling Technology), phosphorylated EGFR and total EGFR, CD44, and Tubulin (Sigma).
  • Antibodies were typically incubated overnight at 4° C. with gentle shake, followed by washes and incubation with the appropriate HRP-conjugated secondary antibodies.
  • Membranes were incubated with chemiluminescent substrate for 5 min at room temperature (SuperSignal West Femto, Thermo Scientific).
  • the chemiluminescent images were acquired with a C-DiGit Imaging System (LI-COR Biosciences).
  • the relative density of the chemiluminescent bands were quantified via Image Studio Digits from LICOR.
  • the half inhibitory concentration (IC 50 ) value is calculated using non-linear regression analysis through GraphPad Prism software (GraphPad, Inc., San Diego, Calif.).
  • Karpas-299 cells were maintained in RPMI medium supplemented with 10% fetal bovine serum and antibiotics. Five hundred thousand cells per well were seeded in 12-well plate and various concentration of compounds were introduced and incubated for 48 hrs. The cells were then collected and lysed in a lysis buffer (20 mM HEPES, 150 mM NaCl, 10 mM KCl, 5 mM EDTA, 1% NP40) supplemented with Halt protease and phosphatase inhibitors (Thermo Scientific).
  • caspase assays approximately 20 i .t.g of cell lysate were incubated with 20 ⁇ l of caspase-3 glo reagent (Promega), and the enzyme activity was measured by the release of luminescence after 20 min incubation at 37° C.
  • caspase-3 glo reagent Promega
  • the enzyme activity was measured by the release of luminescence after 20 min incubation at 37° C.
  • cell lysate were boiled and analyzed by SDS-PAGE/immunoblotting using anti-caspase-3 (Cell Signaling Technology), anti-PARP (Cell Signaling Technology), or anti-actin (Cell Signaling Technology) antibodies.
  • Antibodies were typically incubated overnight at 4° C. with gentle shake, followed by washes and incubation with the appropriate HRP-conjugated secondary antibodies.
  • Membranes were incubated with chemiluminescent substrate for 5 min at room temperature (SuperSignal West Femto, Thermo Scientific), and the chemiluminescent images were obtained with a C-DiGit Imaging System (LI-COR Biosciences).
  • HT1080 or HCC78 cells in RPMI medium supplemented with 10% fetal bovine serum and antibiotics were seeded in 24-well plate. After 12-24 hours, confluent cell monolayers were gently scraped with a sterile pipette tip to form a scratch. The plates were washed with fresh medium, and the cells were incubated with medium alone or medium containing various concentration of compounds. After 12-24 hours, the plates were examined and recorded by an EVOS FL microscopy (Life Technology) to monitor resealing of the cell monolayer.
  • EVOS FL microscopy Life Technology
  • Cell lines were cultured using standard techniques in DMEM or RPMI-1640 medium (Corning, Inc) with 10% fetal bovine serum (Thermo Fisher Scientific, Inc) at 37° C. in a humidified atmosphere with 5% CO 2 .
  • DMEM or RPMI-1640 medium Corning, Inc
  • 10% fetal bovine serum Thermo Fisher Scientific, Inc
  • implantation cells were harvested and pelleted by centrifugation at 250g for 2 minutes. Cells were washed once and resuspended in serum-free medium, with 50% matrigel (v/v) as needed.
  • mice and SCID/Beige mice Female athymic nude mice and SCID/Beige mice (5-8 weeks of age) were obtained from Charles River Laboratory and were housed in Innovive IVC disposable cages on HEPA filtered ventilated racks with ad libitum access to rodent chow and water. Five million cells in 100 ⁇ L serum-free medium were implanted subcutaneously in the right flank region of the mouse. Karpas299 cells were implanted into the SCID/Beige mice. KM12 cells, NIH3T3 EML4-ALK wild type mutant cells, and NIH3T3 SCD4-ROS1 wild type mutant cells were implanted into the athymic nude mice, respectively.
  • mice were implanted with 50% matrigel (Corning, Inc) in the medium. Tumor size and body weight were measured on designated days. Tumor size was measured with an electronic caliper and tumor volume was calculated as the product of length*width 2 *0.5. Mice were randomized by tumor size into treatment groups when tumor volume reached about 100-200 mm 3 and Compound 1 were administered orally (BID) at determined dosage.
  • BID orally
  • mice and SCID/Beige mice Female athymic nude mice and SCID/Beige mice (5-8 weeks of age) were obtained from Charles River Laboratory and were housed in Innovive IVC disposable cages on HEPA filtered ventilated racks with ad libitum access to rodent chow and water. Five million cells in 100 ⁇ L serum-free medium were implanted subcutaneously in the right flank region of the mouse. Ba/F3 EML4-ALK wild type and G1202R mutant cells, Ba/F3 CD74-ROS1 wild type and G2032R mutant cells were implanted into the SCID/Beige mice.
  • NIH3T3 CD74-ROS1 G2032 mutant cells NIH3T3 LMNA-TRKA wild type and G595R mutant cells were implanted into the athymic nude mice, respectively. All models were implanted with 50% matrigel (Corning, Inc) in the medium. Tumor size and body weight were measured on designated days. Tumor size was measured with an electronic caliper and tumor volume was calculated as the product of length*width 2 *0.5. Mice were randomized by tumor size into treatment groups when tumor volume reached about 100-200 mm 3 and Compound 1 were administered orally (BID) at determined dosage.
  • BID orally
  • mice bearing xenograft tumors were humanely euthanized and tumors were resected and snap frozen in liquid nitrogen and stored at ⁇ 80° C. Frozen tumor samples were processed at 4° C. in 1 ⁇ Cell Lysis Buffer (Cell Signaling Technologies) to extract proteins. SDS loading samples were prepared by adding one volume of 4 ⁇ LDS Sample Buffer (Life Technologies, Inc) to three volumes of protein lysate. Tumor SDS protein samples were processed by SDS-PAGE and immunoblotted with rabbit anti-phosphorylated ALK and mouse anti-actin antibodies (Cell Signaling Technologies). The signals from immunoblot were detected by C-DiGit Blot Scanner from LI-COR and the signal intensity were quantified using the Image Studio Digit software (LI-COR).
  • the kinase binding affinity of Compound 1 was screened at DiscoveRx in a panel of 460 kinases followed by the determination of the K d for the hits.
  • Compound 1 demonstrated strong binding affinities with ALK, ROS1, TRKA/B/C, JAK2, SRC, FAK and ARKS (Table 1).
  • the enzymatic kinase inhibition activities of Compound 1 at 10 ⁇ M ATP concentration were determined at Reaction Biology, and the results of IC 50 were summarized in Table 1.
  • the kinase binding affinity of Compound 1 was screened at DiscoveRx in a panel of 460 kinases.
  • the kinase hits of compound 1 were further confired in the recombined enzymatic kinase inhibition assay using 10 ⁇ M ATP concentration at Reaction Biology, and the IC 50 results were summarized in Table 1b.
  • Compound 1 was evaluated against ALK resistant mutations in enzymatic kinase assays with 10 ⁇ M ATP concentration at Reaction Biology, Inc. The results were summarized in Table 2. Potent inhibition by Compound 1 was observed among wild type and mutant ALKs and ROS1s.
  • Compound 1 Potently Inhibited Cell Proliferation in Primary Cell Lines with Oncogenic Fusion or Mutated Genes of ALK, ROS1, TRKA or JAK2
  • ALK fusions are major malignancy drivers in multiple cancer types and cancer cell lines, including lymphoma cell line Karpas-299 harboring NPM-ALK fusion gene, non-small cell lung cancer cell line H2228 harboring EML4-ALK fusion gene, non-small cell lung cancer cell line HCC78 harboring SLC34A2-ROS1 fusion gene, colorectal cancer cell line KM12 harboring TPM3-TRKA fusion gene, and leukemia cell line SET-2 harboring JAK2 V617F mutation.
  • the anti-proliferating activities of Compound 1 on these cell lines were evaluated and the results were summarized in Table 3.
  • Ba/F3 cells engineered to express wild type EML4-ALK fusion gene and mutant EML4-ALKs.
  • the growth of Ba/F3 cells is dependent on interleukin-s (IL-3).
  • IL-3 interleukin-s
  • IL-3 interleukin-s
  • Compound 1 potently inhibited the cell proliferation of various Ba/F3 cell lines with engineered expression of wild and mutant EML4-ALKs, and the results were summarized in Table 3.
  • Ba/F3 cells were engineered to express the oncogenic SDC4-ROS1, SDC4-ROS1 G2032R , CD74-ROS 1, and CD74-ROS1 G2032R fusion genes, respectively.
  • the engineered Ba/F3 cells with fusion ROS1 genes were used to examine the inhibiting activity of Compound 1 on wild and mutant ROS1 fusion genes.
  • the results of cell growth inhibition were summarized in Table 3.
  • Ba/F3 cells were engineered to express the oncogenic CD74-ROS1 L2026M and CD74-ROS1 D2033N fusion genes, respectively.
  • the engineered Ba/F3 cells with fusion ROS1 genes were used to examine the inhibiting activity of Compound 1.
  • the results of cell growth inhibition were summarized in Table 3.
  • Ba/F3 cells were engineered to express the oncogenic LMNA-TRKA and LMNA-TRKA G595R fusion genes, respectively.
  • the engineered Ba/F3 cells with fusion TRKA genes were used to examine the inhibiting activity of Compound 1.
  • the results of cell growth inhibition were summarized in Table 3.
  • Ba/F3 cells were engineered to express the oncogenic TEL-TRKB (also named as ETV6-TRKB) and TEL-TRKB G639R fusion genes, respectively.
  • the engineered Ba/F3 cells with fusion TRKB genes were used to examine the inhibiting activity of Compound ls. The results of cell growth inhibition were summarized in Table 3.
  • Ba/F3 cells were engineered to express the oncogenic TEL-TRKC (also named as ETV6-TRKC), and TEL-TRKC G623R fusion genes, respectively.
  • the engineered Ba/F3 cells with fusion TRKC genes were used to examine the inhibiting activity of Compound 1. The results of cell growth inhibition were summarized in Table 3.
  • Compound 1 caused suppression of ALK autophosphorylation as well as the downstream STAT3 and AKT phosphorylation at IC 50 s of around 1-3 nM in Karpas-299 cell line, which harbors NPM-ALK fusion gene.
  • Compound 1 inhibited ROS1 autophosphorylation as well as the downstream STAT3 and AKT phosphorylation at IC 50 s around 1-3 nM in HCC78 cell line, which harbors SLC34A2-ROS1 fusion gene.
  • Compound 1 inhibited TRKA autophosphorylation as well as the downstream AKT and ERK phosphorylation at IC 50 s around 0.3 nM in KM12 cell line, which harbors TPM3-TRKA fusion gene.
  • Compound 1 inhibited STATS phosphorylation at IC 50 around 158 nM in SET-2 cell line, which harbors JAK2 V617F mutation.
  • Compound 1 inhibited autophosphorylation of ALK with IC 50 of 20-30 nM in Ba/F3 engineered stable cell lines encoding wild type or G1202R mutant EML4-ALK vl fusion genes.
  • Compound 1 inhibited autophosphorylation of ROS1 in Ba/F3 engineered stable cell lines encoding wild type or G2032R mutant CD74-ROS1 or SDC4-ROS1 fusion genes.
  • the pharmacodynamic inhibiting activity of Compound 1 on TRKA, TRKB, TRKC, and FAK signaling in cells was evaluated, and the results were summarized in Table 4a.
  • Compound 1 inhibited FAK phosphorylation as well as the SRC substrate paxillin phosphorylation at IC 50 around 103 nM in NCI-H2228 cell line, which harbors EML4-ALK fusion gene with upregulated SRC and FAK signaling.
  • Compound 1 inhibited autophosphorylation of ROS1 in Ba/F3 engineered stable cell lines encoding L2026M, or D2033N mutant CD74-ROS1 fusion genes.
  • Compound 1 inhibited autophosphorylation of TRKA in NIH3T3 engineered stable cell lines encoding wild type or G595R mutant LMNA-TRKA fusion genes.
  • Compound 1 inhibited autophosphorylation of TRKB in NIH3T3 engineered stable cell lines encoding wild type or G639R mutant TEL-TRKB, also named as ETV6-TRKB fusion genes.
  • Compound 1 increased the number of apoptotic Karpas-299 cells. After 48 hours treatment with Compound 1, Karpas-299 cells were lysed and evaluated with the caspase 3/7 cleavage caspase 3 glo assay or PARP in western blotting assay. The results were presented in FIG. 1 .
  • CD44 is a biomarker of cancer stemness. A high expression level of CD44 was discovered in H2228 cells. Compound 1 suppressed CD44 expression level in H2228 cells in a concentration-dependent manner after 48 hours treatment as shown in FIG. 4 , indicating that compound 1 has the potential to inhibit cancer stemness.
  • Compound 1 The antitumor efficacy of Compound 1 was evaluated in several tumor xenograft models representing cancer populations in which dysregulation of ALK, ROS1 or TRKA is implicated.
  • the NPM-ALK fusion gene in Karpas 299 cells is proved as the driver for tumor growth.
  • SCID/Beige mice bearing Karpas 299 tumors (at the average tumor size of 160 mm 3 ) were dosed with Compound 1 orally BID for seven days ( FIG. 5 ).
  • the control group of mice were given vehicle only.
  • Tumor volume (TMV) was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 5 .
  • An * denotes that the mean TMVs are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • Tumor growth inhibition was calculated as 100%* ⁇ 1 ⁇ [(TMV Treated Last Day of Treatment ⁇ TMV Treated First Day of Treatment )/(TMV Control on Last Day of Treatment ⁇ TMV Control on First Day of Treatment )] ⁇ when TMV Treated Last Day of Treatment ⁇ TMV Treated First Day of Treatment .
  • tumor regression was calculated as 100%*(1 ⁇ TMV Treated Last Day of Treatment /TMV Treated First Day of Treatment ).
  • Compound 1 demonstrated the ability to inhibit tumor growth at 59% and 94% at the dosages of 15 mg/kg and 50 mg/kg BID, respectively.
  • 4 of 8 mice in the 50 mg/kg treatment group exhibited tumor regression. Body weight of the mice were measured on the designated days of the mice and no body weight loss was observed in the Compound 1 treatment groups ( FIG. 6 ).
  • Athymic nude mice bearing the NIH3T3 EML4-ALK WT tumors were dosed with Compound 1 orally BID for 12 days ( FIG. 7 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown as mean ⁇ sem in FIG. 7 .
  • An * denotes that the mean tumor volumes are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • treatment of Compound 1 at 50 mg/kg/BID resulted in 41% tumor regression, with 8 of 8 mice exhibited tumor regression.
  • Compound 1 at the dosage of 15 mg/kg/BID demonstrated the ability to inhibit tumor growth at a TGI of 90%, with 2 of 8 mice exhibited tumor regression. Body weight of the mice were measured on the designated days of the mice and no body weight loss was observed in the Compound 1 treatment groups ( FIG. 8 ).
  • the SDC4-ROS1 fusion gene is considered as the driver for tumor progression in NSCLC.
  • Athymic nude mice bearing the NIH3T3 SDC4-ROS1 WT tumors (at the average tumor size of 100 mm 3 ) were dosed with Compound 1 orally BID for 26 days ( FIG. 9 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 9 .
  • An * denotes that the mean tumor volumes are significantly less in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • the aberrant activity of the TPM3-TRKA is considered as the underlying driver for tumor growth in the KM12 model.
  • Athymic nude mice bearing the KM12 tumors (at the average tumor size of 100 mm 3 ) were dosed with Compound 1 orally BID for seven days ( FIG. 10 ).
  • Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 10 .
  • An * denotes that the mean tumor volumes are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • Compound 1 at the dosage of 15 mg/kg and 75 mg/kg resulted in 13% and 11% tumor regression, respectively.
  • 8 of 8 mice exhibited tumor regression.
  • In the 75 mg/kg group 5 of 8 mice exhibited tumor regression. No loss of body weight were observed in mice treated with Compound 1 compared to those with vehicle control ( FIG. 11 ).
  • Karpas 299 tumors were harvested at either 3 hour or 12 hour after last dose of Compound 1 in a repeated dosing study (15 mg/kg or 50 mg/kg, BID for seven 7 days).
  • the level of ALK phosphorylation was determined by immunoblotting combined with signal quantification by the Image Studio Digit Software.
  • the ability of Compound 1 to inhibit ALK phosphorylation was illustrated in FIG. 12 .
  • ALK phosphorylation was reduced to ⁇ 5% of the control level at 3 hours post oral administration of Compound 1 and was maintained at about 10% of the control level at 12 hours post dosing. This level of phosphorylation inhibition corresponds to 94% TGI.
  • ALK phosphorylation was reduced to ⁇ 10% of the control level at 3 hours post oral administration and was maintained at about 21% of the control level at 12 hours post dosing. This level of ALK phosphorylation corresponds to 59% of TGI.
  • Athymic nude mice bearing the KM12 tumors were dosed with Compound 1 orally BID for seven days at dosages equal or lower than 15 mg/kg for seven days ( FIG. 13 ).
  • Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 13 .
  • An * denotes that the mean tumor volumes are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • Compound 1 inhibited tumor growth in a dose-dependent manner.
  • Compound 1 at the dosage of 3 mg/kg demonstrated the ability to inhibit tumor growth at a TGI of 91%, with 2 of 10 mice exhibiting tumor regression.
  • Compound 1 at the dosage of 1 mg/kg demonstrated the ability to inhibit tumor growth at a TGI of 77%. No loss of body weight were observed in mice treated with Compound 1 compared to those with vehicle control ( FIG. 14 ).
  • SCID/Beige mice bearing the Ba/F3 EML4-ALK WT tumors were dosed with Compound 1 orally BID for 14 days ( FIG. 15 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown as mean ⁇ sem in FIG. 15 .
  • An * denotes that the mean tumor volumes are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • treatment with Compound 1 at 75 mg/kg resulted in 54% tumor regression, with 6 of 8 mice exhibiting tumor regression.
  • Compound 1 at the dosage of 15 mg/kg/BID demonstrated the ability to inhibit tumor growth at a TGI of 74%.
  • Body weight of the mice were measured on the designated days and no body weight loss was observed in the Compound 1 treatment groups ( FIG. 16 ).
  • SCID/Beige mice bearing the Ba/F3 EML4-ALK G1202R tumors were dosed with Compound 1 orally BID for 17 days ( FIG. 17 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown as mean ⁇ sem in FIG. 17 .
  • An * denotes that the mean tumor volumes are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • the CD74-ROS1 fusion gene is considered as one of the drivers for tumor progression in NSCLC.
  • SCID/Beige mice bearing the Ba/F3 CD74-ROS1 WT tumors (at the average tumor size of 200 mm 3 ) were dosed with Compound 1 orally BID for 12 days ( FIG. 19 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 19 .
  • An * denotes that the mean tumor volumes are significantly less in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • SCID/Beige mice bearing the Ba/F3 CD74-ROS1 G2032R tumors were dosed with Compound 1 orally BID for 11 days ( FIG. 21 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 21 .
  • An * denotes that the mean tumor volumes are significantly less in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • the LMNA-TRKA fusion gene is considered as one of the drivers for tumor progression in colorectal cancer.
  • Athymic nude mice bearing the NIH3T3 LMNA-TRKA WT tumors (at the average tumor size of 240 mm 3 ) were dosed with Compound 1 orally BID for five days ( FIG. 23 ).
  • Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 23 .
  • An * denotes that the mean tumor volumes are significantly lower in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
  • Athymic nude mice bearing the NIH3T3 LMNA-TRKA G595R tumors (at the average tumor size of 230 mm 3 ) were dosed with Compound 1 orally BID for five days ( FIG. 25 ).
  • the control group of mice were given vehicle only. Tumor volume was measured by caliper on the indicated days and is shown at mean ⁇ sem in FIG. 25 .
  • An * denotes that the mean tumor volumes are significantly less in the treated group compared to that of the control group (p ⁇ 0.05) as determined by two-way repeated ANOVA followed by post hoc analysis.
US15/745,915 2015-07-21 2016-07-20 Chiral diaryl macrocycles and uses thereof Abandoned US20180325901A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/745,915 US20180325901A1 (en) 2015-07-21 2016-07-20 Chiral diaryl macrocycles and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562195081P 2015-07-21 2015-07-21
US201662302231P 2016-03-02 2016-03-02
US15/745,915 US20180325901A1 (en) 2015-07-21 2016-07-20 Chiral diaryl macrocycles and uses thereof
PCT/US2016/043132 WO2017015367A1 (fr) 2015-07-21 2016-07-20 Macrocycles diaryles chiraux et leurs utilisations

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/043132 A-371-Of-International WO2017015367A1 (fr) 2015-07-21 2016-07-20 Macrocycles diaryles chiraux et leurs utilisations

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/998,763 Continuation US11452725B2 (en) 2015-07-21 2020-08-20 Chiral diaryl macrocycles and uses thereof

Publications (1)

Publication Number Publication Date
US20180325901A1 true US20180325901A1 (en) 2018-11-15

Family

ID=57834656

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/745,915 Abandoned US20180325901A1 (en) 2015-07-21 2016-07-20 Chiral diaryl macrocycles and uses thereof
US16/998,763 Active 2036-07-24 US11452725B2 (en) 2015-07-21 2020-08-20 Chiral diaryl macrocycles and uses thereof
US17/819,858 Pending US20230346786A1 (en) 2015-07-21 2022-08-15 Chiral diaryl macrocycles and uses thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/998,763 Active 2036-07-24 US11452725B2 (en) 2015-07-21 2020-08-20 Chiral diaryl macrocycles and uses thereof
US17/819,858 Pending US20230346786A1 (en) 2015-07-21 2022-08-15 Chiral diaryl macrocycles and uses thereof

Country Status (16)

Country Link
US (3) US20180325901A1 (fr)
EP (2) EP3325488B1 (fr)
JP (1) JP6817287B2 (fr)
KR (1) KR20180033194A (fr)
CN (1) CN108026109B (fr)
AU (1) AU2016296878B2 (fr)
BR (1) BR112018001065A2 (fr)
CA (1) CA2992324A1 (fr)
DK (1) DK3325488T3 (fr)
ES (1) ES2818528T3 (fr)
HK (1) HK1254606A1 (fr)
IL (1) IL256868B (fr)
MX (1) MX2018000718A (fr)
RU (2) RU2728579C2 (fr)
WO (1) WO2017015367A1 (fr)
ZA (1) ZA201800416B (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10618912B2 (en) 2014-01-24 2020-04-14 Turning Point Therapeutics, Inc. Diaryl macrocycles as modulators of protein kinases
US10689400B2 (en) 2016-07-28 2020-06-23 Turning Point Therapeutics, Inc. Macrocycle kinase inhibitors
US10745416B2 (en) 2017-12-19 2020-08-18 Turning Point Therapeutics, Inc. Macrocyclic compounds for treating disease
US11008337B2 (en) 2015-07-02 2021-05-18 Turning Point Therapeutics, Inc. Chiral diaryl macrocycles as modulators of protein kinases
WO2021108672A1 (fr) * 2019-11-27 2021-06-03 Turning Point Therapeutics, Inc. Polythérapie impliquant des composés macrocycliques de diaryle
US11155563B2 (en) 2017-07-28 2021-10-26 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof
US11291667B2 (en) 2017-01-25 2022-04-05 Turning Point Therapeutics, Inc. Combination therapy involving diaryl macrocyclic compounds
US11452725B2 (en) 2015-07-21 2022-09-27 Turning Point Therapeutics, Inc. Chiral diaryl macrocycles and uses thereof
US11731956B2 (en) 2018-10-22 2023-08-22 Alumis Inc. Substituted 1,2,4-triazoles as intermediates in the synthesis of TYK2 inhibitors
US11981684B2 (en) 2022-02-22 2024-05-14 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113354653A (zh) * 2015-07-06 2021-09-07 特普医药公司 二芳基大环多晶型物及其制备方法
CN107267458B (zh) * 2017-07-06 2019-10-29 中南大学湘雅二医院 一种耐克唑替尼人的非小细胞肺癌细胞株h3122-cr23及其应用
CN109516999B (zh) * 2017-11-01 2021-08-17 郑州泰基鸿诺医药股份有限公司 用作蛋白质激酶调节剂的化合物及其应用
TW201927790A (zh) 2017-11-10 2019-07-16 美商安捷克斯製藥公司 作為trk激酶抑制劑之巨環化合物及其用途
JP2021519297A (ja) * 2018-03-28 2021-08-10 フォチョン・ファーマシューティカルズ・リミテッドFochon Pharmaceuticals, Ltd. Trkキナーゼ阻害剤としての大環状化合物
JP7092405B2 (ja) * 2018-04-16 2022-06-28 深▲チェン▼市塔吉瑞生物医薬有限公司 キナーゼ活性を阻害するためのジ(ヘテロ)アリール大環状化合物
WO2019210835A1 (fr) * 2018-05-04 2019-11-07 正大天晴药业集团股份有限公司 Composé macrocyclique de diaryle en tant que modulateur de protéine kinase
CN111171049B (zh) * 2018-11-09 2021-06-04 山东轩竹医药科技有限公司 酪氨酸激酶抑制剂及其用途
JP2022537385A (ja) * 2019-06-19 2022-08-25 ターニング・ポイント・セラピューティクス・インコーポレイテッド 大環状キナーゼ阻害剤の多形
CN111689883B (zh) * 2020-05-22 2022-02-11 上海应用技术大学 一种二芳基砜类化合物的合成方法
EP4163283A1 (fr) * 2020-06-04 2023-04-12 Scinnohub Pharmaceutical Co., Ltd Composé ayant une structure macrocyclique et son utilisation

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT690843E (pt) 1993-03-25 2001-01-31 Upjohn Co Derivados de indolo substituidos com formilo ou ciano possuindo actividade dopaminergica
CN1050844C (zh) * 1993-12-07 2000-03-29 伊莱利利公司 蛋白激酶c抑制剂
RS49779B (sr) 1998-01-12 2008-06-05 Glaxo Group Limited, Biciklična heteroaromatična jedinjenja kao inhibitori protein tirozin kinaze
ATE301661T1 (de) * 2000-12-08 2005-08-15 Ortho Mcneil Pharm Inc Makroheterocyclische verbindungen als kinase inhibitoren
EP2065380A1 (fr) * 2007-08-22 2009-06-03 F.Hoffmann-La Roche Ag Dérivés de pyridoneamide comme inhibiteurs de focal adhesion kinase (FAK) et leur utilisation pour le traitement du cancer
CA2735420C (fr) * 2008-09-08 2016-06-28 Merck Patent Gmbh Pyrimidines macrocycliques en tant qu'inhibiteurs de proteine kinase
CA2738026C (fr) 2008-09-22 2017-01-24 Array Biopharma Inc. Composes imidazo[1,2b]pyridazine substitues comme inhibiteurs de kinases trk
DK3106463T6 (da) 2008-10-22 2020-02-24 Array Biopharma Inc Pyrazolo[1,5-]pyrimidinforbindelse som trk-kinasehæmmer
PE20131197A1 (es) 2008-10-31 2013-11-06 Genentech Inc Compuestos de pirazolopirimidina como inhibidores de jak y composiciones farmaceuticas que los contienen
AR077468A1 (es) 2009-07-09 2011-08-31 Array Biopharma Inc Compuestos de pirazolo (1,5 -a) pirimidina sustituidos como inhibidores de trk- quinasa
ES2446720T3 (es) 2009-10-13 2014-03-10 Elanco Animal Health Ireland Limited Inhibidores de la integrasa macrocíclica
SA111320200B1 (ar) 2010-02-17 2014-02-16 ديبيوفارم اس ايه مركبات ثنائية الحلقة واستخداماتها كمثبطات c-src/jak مزدوجة
HUE035337T2 (en) * 2010-05-20 2018-05-02 Array Biopharma Inc Macrocyclic compounds as TRK kinase inhibitors
US8637516B2 (en) 2010-09-09 2014-01-28 Irm Llc Compounds and compositions as TRK inhibitors
EP2508607A1 (fr) 2011-04-07 2012-10-10 Helmholtz-Zentrum für Infektionsforschung GmbH Medicament for liver regeneration and for treatment of liver failure
WO2013001310A1 (fr) * 2011-06-30 2013-01-03 Centro Nacional De Investigaciones Oncológicas (Cnio) Composés macrocycliques et leur utilisation en tant qu'inhibiteurs de cdk8
KR20140059236A (ko) * 2011-08-19 2014-05-15 머크 샤프 앤드 돔 코포레이션 Hcv 프로테아제 억제제의 결정 형태
WO2013045653A1 (fr) 2011-09-30 2013-04-04 Oncodesign S.A. Inhibiteurs de kinase flt3 macrocycliques
BR112014022106B1 (pt) 2012-03-06 2022-08-02 Pfizer Inc Derivados macrocíclicos, combinação, composição e usos na fabricação de um medicamento para o tratamento de doenças
CA2866164C (fr) 2012-03-09 2020-07-07 Lexicon Pharmaceuticals, Inc. Composes a base d'imidazo [1, 2-b] pyridazine, compositions les comprenant et utilisations de ceux-ci
PT2834243T (pt) 2012-03-09 2018-08-01 Lexicon Pharmaceuticals Inc Compostos à base de pirazolo[1,5-a]pirimidina, composições que os compreendem e métodos para a sua utilização
GB201205669D0 (en) 2012-03-30 2012-05-16 Agency Science Tech & Res Bicyclic heterocyclic derivatives as mnk2 and mnk2 modulators and uses thereof
EA031863B1 (ru) * 2014-01-24 2019-03-29 ТиПи ТЕРАПЬЮТИКС, ИНК. Диарильные макроциклы в качестве модуляторов протеинкиназ
CN107735399B (zh) 2015-07-02 2021-01-26 特普医药公司 作为蛋白质激酶的调节剂的手性二芳基大环
CN113354653A (zh) * 2015-07-06 2021-09-07 特普医药公司 二芳基大环多晶型物及其制备方法
BR112018001065A2 (pt) 2015-07-21 2018-09-11 Tp Therapeutics, Inc. macrociclos diarila quirais e usos dos mesmos
TW201815799A (zh) 2016-07-28 2018-05-01 美商Tp生物醫藥公司 巨環激酶抑制劑
TWI808958B (zh) 2017-01-25 2023-07-21 美商特普醫葯公司 涉及二芳基巨環化合物之組合療法
KR102645316B1 (ko) 2017-07-28 2024-03-07 터닝 포인트 테라퓨틱스, 인크. 거대환형 화합물 및 이의 용도

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10618912B2 (en) 2014-01-24 2020-04-14 Turning Point Therapeutics, Inc. Diaryl macrocycles as modulators of protein kinases
US11008337B2 (en) 2015-07-02 2021-05-18 Turning Point Therapeutics, Inc. Chiral diaryl macrocycles as modulators of protein kinases
US11452725B2 (en) 2015-07-21 2022-09-27 Turning Point Therapeutics, Inc. Chiral diaryl macrocycles and uses thereof
US10689400B2 (en) 2016-07-28 2020-06-23 Turning Point Therapeutics, Inc. Macrocycle kinase inhibitors
US11291667B2 (en) 2017-01-25 2022-04-05 Turning Point Therapeutics, Inc. Combination therapy involving diaryl macrocyclic compounds
US11155563B2 (en) 2017-07-28 2021-10-26 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof
US11286264B2 (en) 2017-07-28 2022-03-29 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof
US10745416B2 (en) 2017-12-19 2020-08-18 Turning Point Therapeutics, Inc. Macrocyclic compounds for treating disease
US11286265B2 (en) 2017-12-19 2022-03-29 Turning Point Therapeutics, Inc. Macrocyclic compounds for treating disease
US11731956B2 (en) 2018-10-22 2023-08-22 Alumis Inc. Substituted 1,2,4-triazoles as intermediates in the synthesis of TYK2 inhibitors
WO2021108672A1 (fr) * 2019-11-27 2021-06-03 Turning Point Therapeutics, Inc. Polythérapie impliquant des composés macrocycliques de diaryle
US11666574B2 (en) 2019-11-27 2023-06-06 Turning Point Therapeutics, Inc. Combination therapy involving diaryl macrocyclic compounds
US11981684B2 (en) 2022-02-22 2024-05-14 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof

Also Published As

Publication number Publication date
KR20180033194A (ko) 2018-04-02
DK3325488T3 (da) 2020-09-14
RU2018106245A3 (fr) 2019-12-26
IL256868A (en) 2018-03-29
US20230346786A1 (en) 2023-11-02
JP6817287B2 (ja) 2021-01-20
BR112018001065A2 (pt) 2018-09-11
US11452725B2 (en) 2022-09-27
ES2818528T3 (es) 2021-04-13
CA2992324A1 (fr) 2017-01-26
EP3325488B1 (fr) 2020-06-24
CN108026109A (zh) 2018-05-11
HK1254606A1 (zh) 2019-07-26
EP3733187A1 (fr) 2020-11-04
CN108026109B (zh) 2021-03-02
MX2018000718A (es) 2020-01-20
RU2020123800A (ru) 2020-10-02
IL256868B (en) 2021-03-25
JP2018524382A (ja) 2018-08-30
AU2016296878B2 (en) 2020-12-17
EP3325488A1 (fr) 2018-05-30
RU2728579C2 (ru) 2020-07-30
ZA201800416B (en) 2021-07-28
WO2017015367A1 (fr) 2017-01-26
RU2018106245A (ru) 2019-08-21
AU2016296878A1 (en) 2018-01-25
EP3325488A4 (fr) 2019-04-24
US20210030756A1 (en) 2021-02-04

Similar Documents

Publication Publication Date Title
US11452725B2 (en) Chiral diaryl macrocycles and uses thereof
US11291667B2 (en) Combination therapy involving diaryl macrocyclic compounds
US20210214373A1 (en) Macrocyclic compounds and uses thereof
JP2016528288A (ja) 上昇したc反応性タンパク質レベルを有する固形腫瘍の患者における延命効果
KR20140043314A (ko) 이중 오로라 키나제/mek 억제제를 사용한 항암 치료법
CA3163095A1 (fr) Polytherapie impliquant des composes macrocycliques de diaryle
WO2021113339A1 (fr) Macrocycles destinés à être utilisés dans le traitement d'une maladie
CN108473472B (zh) 作为p70s6激酶抑制剂的2-氨基喹唑啉衍生物
EP3364971B1 (fr) Inhibiteurs de pdk1 hétérocycliques destinés à être utilisés pour traiter le cancer
CA3213359A1 (fr) Inhibiteurs d'alk-5 et leurs utilisations
CA3174455A1 (fr) Utilisations therapeutiques de composes macrocycliques
US11981684B2 (en) Macrocyclic compounds and uses thereof
US20230158034A1 (en) Co-treatment with cdk4/6 and cdk2 inhibitors to suppress tumor adaptation to cdk2 inhibitors
RU2784853C2 (ru) Комбинационная терапия с применением диарильных макроциклических соединений
CN117561255A (zh) 作为mek抑制剂的3,4-二氢-2,7-萘啶-1,6(2h,7h)-二酮化合物

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

AS Assignment

Owner name: TURNING POINT THERAPEUTICS, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:TP THERAPEUTICS, INC.;REEL/FRAME:050084/0756

Effective date: 20181126

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STCB Information on status: application discontinuation

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