[go: up one dir, main page]

WO2024039883A2 - Compositions and methods to treat myeloproliferative disorders and myeloproliferative neoplasms - Google Patents

Compositions and methods to treat myeloproliferative disorders and myeloproliferative neoplasms Download PDF

Info

Publication number
WO2024039883A2
WO2024039883A2 PCT/US2023/030634 US2023030634W WO2024039883A2 WO 2024039883 A2 WO2024039883 A2 WO 2024039883A2 US 2023030634 W US2023030634 W US 2023030634W WO 2024039883 A2 WO2024039883 A2 WO 2024039883A2
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
alkyl
nhc
cycloalkyl
compound
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.)
Ceased
Application number
PCT/US2023/030634
Other languages
French (fr)
Other versions
WO2024039883A3 (en
Inventor
Stephen Gately
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.)
Translational Drug Development LLC
Original Assignee
Translational Drug Development LLC
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 Translational Drug Development LLC filed Critical Translational Drug Development LLC
Publication of WO2024039883A2 publication Critical patent/WO2024039883A2/en
Publication of WO2024039883A3 publication Critical patent/WO2024039883A3/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4409Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 4, e.g. isoniazid, iproniazid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present application provides the compositions and methods for treating myeloproliferative disorders or neoplasms.
  • the application also provides the methods for preparation of the compositions, the article of manufacture, and the kit thereof.
  • Myeloproliferative disorders or neoplasms are caused by genetic defects in the hematopoietic stem cells, resulting in clonal myeloproliferation, bone marrow fibrosis, and abnormal cytokine expression (Tefferi et al Cancer Res 2022;82:749-63).
  • MPN may be classified into four subtypes: chronic myelogenous leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF).
  • CML chronic myelogenous leukemia
  • PV polycythemia vera
  • ET essential thrombocythemia
  • PMF primary myelofibrosis
  • Treatments of myeloproliferative disorders involve allogeneic stem cell transplant. The transplant procedure is preceded by myeloablative chemotherapy, can lead to severe treatment-related consequences such as graft- versus-host disease, and is limited by performance status
  • JAK2 V617F in Janus kinase 2 or JAK2
  • JAK2 a member of the JAK family of kinases was identified (Baxter et aL, Lancet 365:1054-61, 2005; James et al., Nature 434:1144-8, 2005; Kralovics et al., N. Engl. J. Med. 352:1779-90, 2005; Levine et al., Cancer Cell 7:387-97; 2005).
  • the mutation constitutively activates JAK2 and JAK-STAT signaling, resulting in unrestrained cellular proliferation characteristics of myeloproliferative disorders. It is found in the subtypes of PV, ET, and PMF.
  • jAK2 V517F Vainchenker et al.. Blood 118:1723-35, 2011.
  • JAK inhibitors have been developed for treating myeloproliferative neoplasms, including ruxolitinib (INCB018424) for treating primary myelofibrosis, fedratinib (SAR302503, TG101348) for treating myelofibrosis, and XL019, SB1518 and AZD1480 for treating post-PV/ET myelofibrosis (Sonbol, Ther.
  • CYT387 milotinib
  • N-(cyanomethyl)-4-(2-(4-morpholinophenylamino) pyrimidin-4-yl)benzamide is a different class of JAK inhibitor that provide additional benefits in improving anemia and/or spleen response. It is currently in clinical trials for treating primary myelofibrosis, polycythemia vera (PV), essential thrombocythemia (ET), and post-PV/ET.
  • Fig. 1 depicts gel blot results demonstrating effects on phosphorylated STAT3 in IL-2 activated human peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • Fig. 2 plots the dose dependent inhibitory effect of compound ID#97 on STAT3 phosphorylation in PBMC, compared to KD025 (belumosudil).
  • Fig. 3 show a Western blot of total MLC2 and pMLC2 levels in human HEL 92.1.7 cells after treatment with compound I D#97.
  • Fig. 4 is a bar chart quantifying the effect of compound ID#97 on MLC phosphorylation in human HEL 92.1.7 cells.
  • Fig. 5 is a Western blot of total MLC2 and pMLC2 levels in human UKE-1 Cells after treatment with compound I D#97.
  • Fig. 6 is a bar chart quantifying the effect of compound ID#97 on MLC phosphorylation in human UKE-1 cells.
  • Fig. 7 depicts CFU assay in human HEL-92.1.7 cells with serial dilution of compound ID#97.
  • Fig. 8 depicts CFU assay in human UKE-1 cells with serial dilution of compound ID#97.
  • Fig. 9 is a cell cycle analysis in human HEL-92.1.7 cells.
  • Fig. 10 is a cell cycle analysis in human UKE-1 cells.
  • Fig. 11 shows cell cycle distributions for HEL-92.1.7 cells and UKE-1 cells, after treatment with compound #10 or compound #97.
  • Fig. 12 is a bar graph depicting colony forming unit assay results.
  • Fig. 13 is a summary of the colony forming unit assay results, listing mean CFU +/- SEM.
  • Fig. 14 plots in vivo survival rates after i.v. treatment with compound #97, with or without Ruxolitinib, on HEL-92.1.7 cells.
  • Protein kinases serve to catalyze the phosphorylation of an amino acid side chain in various proteins by the transfer of the y-phosphate of the ATP-Mg2+ complex to said amino acid side chain. These enzymes control the majority of the signaling processes inside cells, thereby governing cell function, growth, differentiation, and destruction (apoptosis) through reversible phosphorylation of the hydroxyl groups of serine, threonine, and tyrosine residues in proteins. Studies have shown that protein kinases are key regulators of many cell functions, including signal transduction, transcriptional regulation, cell motility, and cell division.
  • the protein kinase family of enzymes is typically classified into two main subfamilies: Protein Tyrosine Kinases and Protein Serine/Threonine Kinases, based on the amino acid residue they phosphorylate.
  • the serine/threonine kinases includes cyclic AMP- and cyclic GMP- dependent protein kinases, calcium- and phospholipid-dependent protein kinase, calcium- and calmodulin-dependent protein kinases, casein kinases, cell division cycle protein kinases and others. These kinases are usually cytoplasmic or associated with the particulate fractions of cells, possibly by anchoring proteins.
  • tyrosine kinases phosphorylate tyrosine residues.
  • Tyrosine kinases play an equally important role in cell regulation. These kinases include several receptors for molecules such as growth factors and hormones, including epidermal growth factor receptor, insulin receptor, platelet derived growth factor receptor and others.
  • tyrosine kinases are transmembrane proteins with their receptor domains located on the outside of the cell and their kinase 5 domains on the inside. Much work is also under progress to identify modulators of tyrosine kinases as well.
  • RhoA is a small GTP binding protein that can be activated by several extracellular stimuli such as growth factor, hormones, mechanic stress, osmotic change as well as high concentration of metabolite like glucose. RhoA activation involves GTP binding, conformation alteration, post-translational modification (geranylgeranylation and famesylation) and activation of its intrinsic GTPase activity. Activated RhoA can interact with several effector proteins including ROCKs and transmit signals into cellular cytoplasm and nucleus.
  • ROCK1 and 2 constitute a family of kinases that can be activated by RhoA-GTP complex via physical association. Activated ROCKs phosphorylate several substrates and play important roles in pivotal cellular functions.
  • the substrates for ROCKs include myosin binding subunit of myosin light chain phosphatase (MBS, also named MYPT1), adducin, myosin light chain (MLC), LIM kinase as well as transcription factor FHL.
  • MCS myosin binding subunit of myosin light chain phosphatase
  • MLC myosin light chain
  • LIM kinase as well as transcription factor FHL.
  • RhoA kinase activity of ROCK1 and which in turn phosphorylates MBS.
  • MLCK calciumdependent myosin light chain kinase
  • ROCKs have also been shown to be involved in cellular functions including apoptosis, cell migration, transcriptional activation, fibrosis, cytokinesis, inflammation, and cell proliferation.
  • ROCK plays a critical role in the inhibition of axonal growth by myelin-associated inhibitory factors such as i myelin-associated glycoprotein (MAG).
  • MAG myelin-associated glycoprotein
  • ROCK-induced phosphorylation of substrates such as LIM kinase and myosin light chain phosphatase, resulting in increased contractility of the neuronal actin-myosin system.
  • Inhibitors of ROCKs have been suggested for use in the treatments of a variety of diseases. They include cardiovascular diseases such as hypertension, chronic and congestive heart failure, cardiac hypertrophy, restenosis, chronic renal failure, and atherosclerosis. In addition, because of its muscle relaxing properties, it is also suitable for asthma, male erectile dysfunctions, female sexual dysfunction, and over-active bladder syndrome. ROCK inhibitors have been shown to possess anti-inflammatory properties.
  • ROCK inhibitors could be useful drugs for neuronal regeneration, inducing new axonal growth and axonal rewiring across lesions within the CNS.
  • ROCK inhibitors are therefore likely to be useful for regenerative (recovery) treatment of CNS disorders such as spinal cord injury, acute neuronal injury (stroke, traumatic brain injury), Parkinson's disease, Alzheimer's disease, and other neurodegenerative disorders. Since ROCK inhibitors reduce cell proliferation and cell migration, they could be useful in treating cancer and tumor metastasis. Furthermore, there is evidence suggesting that ROCK inhibitors suppress cytoskeletal rearrangement upon virus invasion, thus they also have potential therapeutic value in antiviral and anti-bacterial applications. ROCK inhibitors may also be useful for the treatment of insulin resistance and diabetes.
  • the aurora family of serine/threonine protein kinases is essential for cell proliferation.
  • the human aurora kinase family consists of three highly homologous kinases (A or "2", B or “1” and C or “3"). During normal cell proliferation, these proteins are involved in chromosome segregation, mitotic spindle function, and cytokinesis. Aurora kinase expression is low in resting cells and peaks during the G2 and mitosis phases of the cell cycle.
  • Several proposed mammalian substrates for Aurora kinases that are important for cell division include histone H3, TPX2, myosin II regulatory light chain, CENP-A, and protein phosphatase 1.
  • Aurora kinases Since the elucidation of their key role in mitotic progression and cell division, Aurora kinases have been closely linked to tumorigenesis. For example, Aurora kinase gene amplification and overexpression has been reported in many cancers. A coding single nucleotide polymorphism (SNP) has been identified that is significantly more frequent in advanced gastric cancer relative to early-stage gastric cancer, and this SNP correlates with elevated kinase activity. Overexpression of Aurora A induces centrosome amplification, aneuploidy, and transformation in rodent fibroblasts. This oncogenic activity is likely due to the generation of chromosome instability. Indeed, there is a strong correlation between Aurora A overexpression and chromosome aneuploidy in breast and gastric cancer.
  • SNP single nucleotide polymorphism
  • Aurora B expression is elevated in cell lines derived from tumors of the colon, breast, lung, melanoma, kidney, ovary, pancreas, CNS, gastric tract and leukemias.
  • increased nuclear expression of Aurora B was observed in high Gleason grade anaplastic prostate cancer tissues relative to low and intermediate grades, and Aurora B expression was accompanied by the phosphorylation of the histone H3 substrate.
  • Aurora kinase inhibition in tumor cells can result in mitotic arrest and apoptosis, these kinases are important targets for cancer therapy. Given the central role of mitosis in the progression of virtually all malignancies, inhibitors of the Aurora kinases therefore are expected to have the potential to block growth of cancers or tumors and have application across a broad range of human cancers.
  • ROCK1 and ROCK2 expression is elevated in patients with essential thrombocythemia (ET), myelofibrosis (MF), and polycythemia vera (PV) compared to healthy individuals (Saleiro et al, Nature Communications (2022) 13:1750).
  • ROCK kinases and aurora kinases are involved in megakaryocyte differentiation and maturation, platelet production, and proplatelet formation and this is implicated in myeloproliferative neoplasms (Yang et al, Exp Hematol. 2017 April ; 48: 32-38)
  • the methods described herein provide a treatment for a myeloproliferative disorder, comprising administering to a patient a therapeutic effective amount of JAK inhibitor and a therapeutic effective amount of a ROCK inhibitor.
  • the JAK inhibitor is selected from the group consisting of ruxolitinib, fedratinib, tofacitinib, baricitinib, lestaurtinib, pacritinib, XL019, AZD1480, INCB039110, LY2784544, BMS911543, NS018, or N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide; or pharmaceutically acceptable salts thereof.
  • the JAK2 inhibitor is ruxolitinib.
  • the JAK inhibitor is a JAK2 inhibitor N-(cyanomethyl)-4-[2-(4-morpholinoanilino)pyrimidin-4-yl]benzamide or a pharmaceutically acceptable salt thereof.
  • the JAK inhibitors are selected from Decernotinib (or VX-509), GLPG0634, or GLPG0788, or a pharmaceutically acceptable salt thereof.
  • the method of treating myeloproliferative diseases further comprises one or more therapeutic agents selected from a spleen tyrosine kinase (SYK) inhibitor, a Bruton's tyrosine kinase (BTK) inhibitor, a bromodomain-containing protein (BRD) inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anticancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof.
  • SYK spleen tyrosine kinase
  • BTK Bruton's tyrosine kinase
  • BBD bromodomain-containing protein
  • chemotherapeutic agent an immunotherapeutic agent
  • a radiotherapeutic agent an anti-neoplastic agent
  • an anticancer agent an anti-proliferation agent
  • Additional methods include one or more therapeutic agent selected from a PI3K (including PI 3Ky, PI3K6, PI3KI 3, and PI3Ka inhibitor, a JAK (including JAK1 and JAK2) inhibitor, a SYK inhibitor, a BTK inhibitor, a BRD (including BRD4 inhibitor), a, BCL2 inhibitor, a LOXL (including LOXL1, LOXL2, LOXL3, LOXL4, or LOXLS) inhibitor, a MMP (including MMP2 and MMP9) inhibitor, a A2B inhibitor, an IDH (including IDH1) inhibitor, an ASK (including ASK1) inhibitor, a TPL2 inhibitor, a DDR (including DDR1 and DDR2) inhibitor, an HDAC inhibitor, a PKC inhibitor, or any combination thereof.
  • a PI3K including PI 3Ky, PI3K6, PI3KI 3, and PI3Ka inhibitor
  • a JAK including JAK1 and JAK2
  • one or more therapeutic agents are selected from an Abl inhibitor, an ACK inhibitor, an A2B inhibitor, an ASK inhibitor, an Aurora kinase inhibitor, a BTK inhibitor, a BRD inhibitor, a c-Kit inhibitor, a c-Met inhibitor, a CAK inhibitor, a CaMK inhibitor, a CDK inhibitor, a CK inhibitor, a DDR inhibitor, an EGFR inhibitor, a FAK inhibitor, a Flt-3 inhibitor, a FYN inhibitor, a GSK inhibitor, a HCK inhibitor, a HDAC inhibitor, an IKK inhibitor, an IDH inhibitor, an IKK inhibitor, a KDR inhibitor, a LCK inhibitor, a LOX inhibitor, a LOXL inhibitor, a LYN inhibitor, a MMP inhibitor, a MEK inhibitor, a MAPK inhibitor, a MDM2 inhibitor, a NEK9 inhibitor, a NPM-ALK inhibitor, a p38 kinase inhibitor, a PDGF inhibitor,
  • the myeloproliferative disorder is selected from the group consisting of polycythemia vera (PV), primary myelofibrosis (PMF), thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), chronic myelogenous leukemia (CML), systemic mastocystosis (SM), chronic neutrophilic leukemia (CNL), myelodysplastic syndrome (MDS) and systemic mast cell disease (SMCD).
  • the myeloproliferative disorder is myelofibrosis (MF).
  • a treatment is provided for patients having myeloproliferative disorder selected from the group consisting of polycythemia vera (PV), primary myelofibrosis (PMF), or essential thrombocythemia (ET).
  • PV polycythemia vera
  • PMF primary myelofibrosis
  • ET essential thrombocythemia
  • the patient has received prior treatment and/or develops disease persistence to treatment of myeloproliferative disorder or has not previously been treated for myeloproliferative disorder.
  • a treatment is provided for patients having diseases selected from diffuse large B- cell lymphoma.
  • a method for decreasing cell viability, decreasing proliferation, or increasing apoptosis comprises contacting cells with an effective amount of JAK inhibitor and an effective amount of PI3K inhibitor.
  • the JAK inhibitor is selected from the group consisting of ruxolitinib, fedratinib, tofacitinib, baricitinib, lestaurtinib, pacritinib, XL019, AZD1480, INCB039110, LY2784544, BMS911543, NS018, or N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide; or pharmaceutically acceptable salts thereof.
  • the method uses cells that are isolated from a subject having myeloproliferative disorder selected from the group consisting of polycythemia vera (PV), primary myelofibrosis (PMF), thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), chronic myelogenous leukemia (CML), systemic mastocystosis (SM), chronic neutrophilic leukemia (CNL), myelodysplastic syndrome (MDS) and systemic mast cell disease (SMCD). Also, the method uses cells that are isolated from a subject having diffuse large B-cell lymphoma (DLBCL).
  • PV polycythemia vera
  • PMF primary myelofibrosis
  • thrombocythemia thrombocythemia
  • ET essential thrombocythemia
  • IMF idiopathic myelofibrosis
  • CML chronic myelogenous leukemia
  • SM chronic
  • a pharmaceutical composition comprising a therapeutically effective amount of JAK inhibitor, a therapeutically effective amount of ROCK inhibitor, and a pharmaceutically acceptable excipient is provided.
  • kits comprising a pharmaceutical composition and a label.
  • the kit contains the pharmaceutical composition that comprises a therapeutically effective amount of JAK inhibitor, a therapeutically effective amount of ROCK inhibitor, and a pharmaceutically acceptable excipient.
  • the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a myeloproliferative disorder.
  • the application provides a JAK2 inhibitor N-(cyanomethyl)-4-[2-(4-morpholinoanilino)pyrimidin-4-yl]benzamide; or a pharmaceutically acceptable salt thereof, which is administered at a dose between 50 to 350 mg; or between 100 to 200 mg.
  • the method of treating myeloproliferative diseases further comprises one or more therapeutic agents selected from a SYK inhibitor, a BTK inhibitor, a BRD inhibitor, a MDM2 inhibitor, a telomerase inhibitor, a BCL2 inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof.
  • the administration of the JAK inhibitor is prior to the administration of the ROCK inhibitor.
  • the administration of the JAK inhibitor is concurrent to the administration of the ROCK inhibitor.
  • the administration of the JAK inhibitor is after the administration of the ROCK inhibitor.
  • the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a hyperproliferative disorder.
  • the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a hyperproliferative disorder.
  • the application provides a ROCK inhibitor for use in a method for treating a hyperproliferative disorder.
  • the application provides a ROCK inhibitor for use in a method fortreating a hyperproliferative disorder.
  • the hyperproliferative disorder is myeloproliferative disorder.
  • the hyperproliferative disorder is cancer.
  • the application provides a ROCK inhibitor for use in treating hyperproliferative disorders or neoplasms, wherein the ROCK inhibitor is administered simultaneously, separately, or sequentially with a ROCK inhibitor.
  • the method of treating hyperproliferative diseases comprising administering a therapeutically effective amount of an Abl inhibitor, an ACK inhibitor, an A2B inhibitor, an ASK inhibitor, an Aurora kinase inhibitor, a BTK inhibitor, a BRD inhibitor, a BCL2 inhibitor, a c-Kit inhibitor, a c-Met inhibitor, a CAK inhibitor, a CaMK inhibitor, a CDK inhibitor, a CK inhibitor, a DDR inhibitor, an EGFR inhibitor, a FAK inhibitor, a Flt-3 inhibitor, a FYN inhibitor, a GSK inhibitor, a HCK inhibitor, a HDAC inhibitor, an IKK inhibitor, an IDH inhibitor, an IKK inhibitor, a JAK inhibitor, a KDR inhibitor, a LCK inhibitor, a LOX inhibitor, a LOXL inhibitor, a LYN inhibitor, a MMP inhibitor, a MEK inhibitor, a MARK inhibitor, a MDM2 inhibitor, a NEK9 inhibitor, a
  • the one or more therapeutic agent is selected from a ROCK inhibitor, a JAK (including JAK1 and JAK2) inhibitor, a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor, a SYK inhibitor, a BTK inhibitor, a BRD (including BRD4 inhibitor), a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti- neoplastic agent, an anti-cancer agent, an anti-proliferation agent, or any combination thereof.
  • the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a myeloproliferative disorder.
  • the application provides a ROCK inhibitor for use in a method for treating a myeloproliferative disorder.
  • the application provides a ROCK inhibitor for use in a method for treating a myeloproliferative disorder.
  • the administration of the JAK inhibitor is prior to the administration of the ROCK inhibitor.
  • the application provides a JAK2 inhibitor N-(cyanomethyl)-4-[2- (4-morpholinoanilino)pyrimidin-4-yl]benzamide; or a pharmaceutically acceptable hydrochloride salt thereof, which is administered at a dose between 100 to 300 mg.
  • the application provides a JAK inhibitor ruxolitinib, or a pharmaceutically acceptable phosphate salt thereof, which is administered at a dose between 15 to 25 mg.
  • the present application provides methods for treating hyperproliferative disorders such as cancers and myeloproliferative disorders in a subject by administering one or more therapeutic agents.
  • the myeloproliferative disorders also referred to as myeloproliferative neoplasms (MPN)
  • MPD myeloproliferative disorders
  • MPN myeloproliferative neoplasms
  • MPN includes, among others, polycythemia vera (PV), primary myelofibrosis, thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis, chronic myelogenous leukemia (CIVIL), systemic mastocystosis, chronic neutrophilic leukemia, myelodysplastic syndrome, and systemic mast cell disease.
  • PV polycythemia vera
  • E essential thrombocythemia
  • CIVIL chronic myelogenous leukemia
  • systemic mastocystosis chronic neutrophilic leukemia
  • myelodysplastic syndrome myelodysplastic syndrome
  • AML acute myeloid leukemia
  • Current MPN therapies aim at providing palliative care over a long period of time.
  • the methods provided herein treat myeloproliferative diseases by administering one or more therapeutic agents for treating myeloproliferative diseases.
  • the methods use or include a single therapeutic agent.
  • the methods use or include a combination of two or more therapeutic agents.
  • a method is provided for treating myeloproliferative diseases by administering a combination of therapeutic agents or small molecule inhibitors that inhibit B-cell receptor (BCR)-mediated signaling, phosphatidylinositol 3-kinase (PI3K)-mediated, Janus kinase (JAK)-mediated signaling pathways, BCL2 inhibitors, MDM2 inhibitors, telomerase inhibitors or any combination thereof.
  • BCR B-cell receptor
  • PI3K phosphatidylinositol 3-kinase
  • JAK Janus kinase
  • a therapeutic agent may be a compound or a biologic molecule (such as DNA, RNA, or protein) that provide desired therapeutic effects when administered to a subject in need thereof (e.g., MPN patients).
  • the therapeutic agent is a compound that inhibits kinase that, directly or indirectly, relates to the disease mechanism or development.
  • enhanced therapeutic effects or variants thereof refer to additional beneficial or synergistic effects to patients that are not observed previously, including fewer and/or reduced symptoms, higher survival rate, prolonged survival time, shorter treatment duration, lower drug dosage, increased molecular and/or cellular responses, and the like.
  • the combination of therapeutic agents or inhibitors may target upstream or downstream components of the same pathway.
  • the combination of therapeutic agents or inhibitors may target different components of dual or multiple pathways. It is hypothesized that the use of a combination of therapeutic agents or inhibitors may enhance therapeutic effects compared to the use of a single therapeutic agent or inhibitor.
  • compositions including pharmaceutical compositions, formulations, or unit dosages
  • articles of manufacture and kits comprising one or more therapeutic agents, including a ROCK inhibitor, a PI3K inhibitor (including a PI3K6 inhibitor), a spleen tyrosine kinase (SYK) inhibitor, a Janus kinase (JAK) inhibitor (including a JAK2 inhibitor), a Bruton's tyrosine kinase (BTK) inhibitor, and a bromodomain containing protein inhibitor (BRD) inhibitor (including a BRD4 inhibitor), a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor.
  • a ROCK inhibitor including a PI3K inhibitor (including a PI3K6 inhibitor), a spleen tyrosine kinase (SYK) inhibitor, a Janus kinase (JAK) inhibitor (including a JAK2 inhibitor), a Bruton's ty
  • one or more therapeutic agent is selected from a PI3K (including PI3Ky, PI3K6, PI3K , PI3Ka, and/or pan-PI3K) inhibitor, a JAK (including JAK1 and/or JAK2) inhibitor, a SYK inhibitor, a BTK inhibitor, a BCL2 inhibitor, an A2B (adenosine A2B receptor) inhibitor, an ACK (activated CDC kinase, including ACK1) inhibitor, an ASK (apoptosis signal-regulating kinase, including ASK1) inhibitor, Aurora kinase, a BRD (bromodomain-containing protein, including BRD4) inhibitor, a BCL2 inhibitor, a CAK (CDK-activating kinase) inhibitor, a CaMK (calmodulindependent protein kinases) inhibitor, a CDK (cyclin-dependent kinases, including CDK1, 2, 3, 4, and/or 6) inhibitor, a CK (
  • the administration of a ROCK inhibitor, and a JAK inhibitor, including N-(cyanomethyl)-4-(2-((4-morpholinophenyl)amino)pyrimidin-4-yl)benza- mide or ruxolitinib, to diseased cells or patients has led to unexpected enhanced therapeutic effects compared to the administration of each kinase inhibitor alone.
  • the unexpected synergistic effects include, but are not limited to, for example, decreased cell viability, increased cell death or apoptosis, decreased inhibition or interference with ROCK signaling pathways, and/or reduction in chemokine (e.g., CCL2, CCL3, CLL4 and CLL22) production, reduced colony formation in diseased cells or patients.
  • unexpected effects may include, but are not limited to, increased inhibition or interference of JAK/STAT (including STAT3 and STAT5), decreased doses or duration of a single agent treatment. Further, the administration of both ROCK and JAK inhibitors unexpectedly restored or increased sensitivity or response of the diseased cells that had developed resistance, or the patients developed disease persistence to prior treatment.
  • the present application provides methods, compositions, kits, and articles of manufacture thereof that use or include one or more therapeutic agents inhibiting one or more targets that relate to, directly or indirectly, cell growth, proliferation, or apoptosis for treating hyperproliferative disorders such as cancers or myeloproliferative neoplasms.
  • the one or more therapeutic agents are compounds or molecules that target a PI3 kinase (PI3K), a spleen tyrosine kinase (SYK), a Janus kinase (JAK), a bromodomain-containing (BRD), a Bruton's tyrosine kinase (BTK), a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor, or any combination thereof, resulting in the inhibition of the target.
  • the therapeutic agent is a ROCK inhibitor that inhibits ROCK1 and ROCK2.
  • the therapeutic agents are a ROCK inhibitor and a JAK2 inhibitor.
  • the JAK inhibitor binds and inhibits one or more members of JAK family, including JAK1, JAK2, and/or JAK3.
  • the JAK inhibitor is N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide, also referred to as CYT0387 or momelotinib, that is a selective inhibitor to JAK2 and JAK1, relative to JAK3.
  • CYT0387 or momelotinib N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide
  • JAK inhibitors include, but are not limited to, ruxolitinib (INCB018424), fedratinib (SAR302503, TG101348), tofacitinib, baricitinib, lestaurtinib, pacritinib (SB1518), XL019, AZD1480, INCB039110, LY2784544, BMS911543, and NS018.
  • Other JAK inhibitors include, but not limited to, Decernotinib (or VX-509), GLPG0634, or GLPG0788, or a pharmaceutically acceptable salt thereof.
  • the compound structures may be named or identified using commonly recognized nomenclature systems and symbols.
  • the compound may be named or identified with common names, systematic or non-systematic names
  • the nomenclature systems and symbols that are commonly recognized in the art of chemistry include, for example, ChemBioDraw Ultra 12.0, Chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (IUPAC).
  • the one or more therapeutic agents include inhibitors that are being used and/or developed to treat various hyperproliferative disorders such as cancer or myeloproliferative neoplasms.
  • Exemplified therapeutic agents include compounds or molecules inhibiting pathways related to BCR, BCL2.
  • Inhibitors of mTOR include temsirolimus, everolimus, ridaforolimus (or deforolimus), OSI-027, AZD2014, CC-223, RADOOl, LY294002, BEZ235, rapamycin, Ku-0063794, or PP242.
  • Inhibitors of AKT include MK-2206, GDC-0068 and GSK795.
  • Inhibitors of MEK includes trametinib, selumetinib, cobimetinib, MEK162, PD-325901, PD-035901, AZD6244, and CI-1040.
  • the application also uses and includes other inhibitors, such as CDK inhibitors (AT-7519, SNS- 032), JNK inhibitors (CC-401), MAPK inhibitors (VX-702, SB203580, SB202190), Raf inhibitors (PLX4720), Tie2 inhibitor (AMG-Tie2-1).
  • CDK inhibitors AT-7519, SNS- 032
  • JNK inhibitors CC-401
  • MAPK inhibitors VX-702, SB203580, SB202190
  • Raf inhibitors PLX4720
  • Tie2 inhibitor AMG-Tie2-1
  • such inhibitors include compounds or agents that inhibit all subclasses (e.g., isoforms or members) of a target (e.g. PI3K alpha
  • the one or more therapeutic agents may be used or combined with a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof.
  • the one or more therapeutic agents are compounds or molecules that is an Abl inhibitor, an ACK inhibitor, an A2B inhibitor, an ASK inhibitor, an Aurora kinase inhibitor, a BTK inhibitor, a BRD inhibitor, a BCL2 inhibitor, a c- Kit inhibitor, a c-Met inhibitor, a CAK inhibitor, a CaMK inhibitor, a CDK inhibitor, a CK inhibitor, a DDR inhibitor, an EGFR inhibitor, a FAK inhibitor, a Flt-3 inhibitor, a FYN inhibitor, a GSK inhibitor, a HCK inhibitor, a HDAC inhibitor, an IKK inhibitor, an IDH inhibitor, an IKK inhibitor, a JAK inhibitor, a KDR inhibitor, a LCK inhibitor, a LOX inhibitor, a LOXL inhibitor, a LYN inhibitor, a MMP inhibitor, a MEK inhibitor, a MARK inhibitor, a MDM2 inhibitor, a NEK9 inhibitor, a NPM- ALK inhibitor, a
  • Chemotherapeutic agents may be categorized by their mechanism of action into, for example, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (floxuridine, capecitabine, and cytarabine); purine analogs, folate antagonists and related inhibitors antiproliferative/antimitotic agents including natural products such as vinca alkaloid (vinblastine, vincristine) and microtubule such as taxane (paclitaxel, docetaxel), vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide); DNA damaging agents (actinomycin, amsacrine, busulfan, carboplatin, chlorambucil, cisplatin, cyclophosphamide, Cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin, iphosphamide,
  • chemotherapeutic agent or “chemotherapeutic” (or “chemotherapy,” in the case of treatment with a chemotherapeutic agent) is meant to encompass any non-proteinaceous (i.e, non-peptidic) chemical compound useful in the treatment of cancer.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN(tm)); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; emylerumines and memylamelamines including alfretamine, triemylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimemylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (articularly cryptophycin 1 and cryptophycin 8); dolastatin
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzi nostatin chromophore and related chromoprotein enediyne antibiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carrninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (AdramycinTM) (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, 6-diazo-5-oxo-L-norle
  • chemotherapeutic agent anti-hormonal agents that act to regulate or inhibit hormone action on tumors
  • anti-estrogens and selective estrogen receptor modulators SERMs
  • SERMs selective estrogen receptor modulators
  • tamoxifen including NolvadexTM
  • raloxifene including NolvadexTM
  • droloxifene 4-hydroxytamoxifen
  • trioxifene keoxifene
  • LY117018 4-hydroxytamoxifen
  • toremifene Fareston®
  • inhibitors of the enzyme aromatase which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate (Megace®), exemestane, formestane, fadrozole, vorozole (Rivisor®), letrozole (Femara®), and anastrozole (Arimidex®)
  • anti-androgens such as flutamide
  • the anti-angiogenic agents include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATIN®, ENDOSTATIN®, suramin, squalamine, tissue inhibitor of metalloproteinase-1, tissue inhibitor of metalloproternase-2, plasminogen activator inhibitor-1, plasminogen activator inbibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulphate (clupeine), sulphated chitin derivatives (prepared from queen crab shells), sulphated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-azetidine-2-carboxylic acid (LACA), cishydroxyproline, d,l-3,4-dehydroproline, thiaproline, .alpha.
  • proline analogs ((l-
  • anti-angiogenesis agents include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: beta-FGF, alpha-FGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-l/Ang-2. See Ferrara N. and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.
  • the anti-fibrotic agents include, but are not limited to, the compounds such as beta- aminoproprionitrile (BAPN), as well as the compounds disclosed in U.S. Pat. No. 4,965,288 to Palfreyman, et al., issued Oct. 23, 1990, entitled "Inhibitors of lysyl oxidase,” relating to inhibitors of lysyl oxidase and their use in the treatment of diseases and conditions associated with the abnormal deposition of collagen; U.S. Pat. No. 4,997,854 to Kagan, et al., issued Mar.
  • BAPN beta- aminoproprionitrile
  • Exemplary anti-fibrotic agents also include the primary amines reacting with the carbonyl group of the active site of the lysyl oxidases, and more particularly those which produce, after binding with the carbonyl, a product stabilized by resonance, such as the following primary amines: emylenemamine, hydrazine, phenylhydrazine, and their derivatives, semicarbazide, and urea derivatives, aminonitriles, such as betaaminopropionitrile (BAPN), or 2-nitroethylamine, unsaturated or saturated haloamines, such as 2-bromo-ethylamine, 2-chloroethylamine, 2-trifluoroethylamine, 3-bromopropylamine, p- halobenzylamines, selenohomocysteine lactone.
  • primary amines reacting with the carbonyl group of the active site of the lysyl oxidases, and more particularly those which produce
  • the anti-fibrotic agents are copper chelating agents, penetrating or not penetrating the cells.
  • Exemplary compounds include indirect inhibitors such compounds blocking the aldehyde derivatives originating from the oxidative deamination of the lysyl and hydroxylysyl residues by the lysyl oxidases, such as the thiolamines, in particular D-penicillamine, or its analogues such as 2-amino-5-mercapto-5-methylhexanoic acid, D-2-amino-3-methyl-3-((2-acetamidoethyl)dithio)butanoic acid, p-2-amino-3-methyl-3-((2- aminoethyl)dithio)butanoic acid, sodium-4-((p-l-dimethyl-2-amino-2-carboxyethyl)dithio) butane sulphurate, 2-acetamidoethyl-2-acetamido
  • the immunotherapeutic agents include and are not limited to therapeutic antibodies suitable for treating patients; such as abagovomab, adecatumumab, afutuzumab, alemtuzumab, altumomab, amatuximab, anatumomab, arcitumomab, bavituximab, bectumomab, bevacizumab, bivatuzumab, blinatumomab, brentuximab, cantuzumab, catumaxomab, cetuximab, citatuzumab, cixutumumab, clivatuzumab, conatumumab, daratumumab, drozitumab, duligotumab, dusigitumab, detumomab, dacetuzumab, dalotuzumab, ecromeximab, elotuzumab
  • the one or more additional therapeutic agent may be an inhibitor to PI3K such as PI3Ky, PI3K6, PI3K0, and PI3Ka, JAK such as JAK1 and JAK2, SYK, BTK, BRD such as BRD4, BCL2 inhibitor, lysyl oxidase protein, lysyl oxidase-like protein (LOXL) such as LOXL1, LOXL2, LOXL3, LOXL4, or LOXLS, matrix metalloprotease (MMP) such as MMP 1-10, adenosine A2B receptor (A2b), isocitrate dehydrogenase (IDH) such as IDH1, apoptosis signal-regulating kinase (ASK) such as ASK1, serine/threonine kinase TPL2, discoidin domain receptor (DDR) such as DDR1 and DDR2, histone deacetylase (HDAC) inhibitor protein
  • the one or more therapeutic agent may be a PI3K (including PI3 Ky, PI3K6, PI3K0, PI3Ka, and/or pan-PI3K) inhibitor, a JAK (including JAK1 and/or JAK2) inhibitor, a SYK inhibitor, a BTK inhibitor, an A2B (adenosine A2B receptor) inhibitor, an ACK (activated CDC kinase, including ACK1) inhibitor, an ASK (apoptosis signal-regulating kinase, including ASK1) inhibitor, Aurora kinase, a BRD (bromodomain-containing protein, including BRD4) inhibitor, a CAK (CDK-activating kinase) inhibitor, a CaMK (calmodulin-dependent protein kinases) inhibitor, a CDK (cyclin-dependent kinases, including CDK1, 2, 3, 4, and/or 6) inhibitor, a CK (casein kinase, including CK
  • the one or more therapeutic agent is: a JAK inhibitor, including but not limited to ruxolitinib, fedratinib, tofacitinib, baricitinib, lestaurtinib, pacritinib, XL019, AZD1480, INCB039110, LY2784544, BMS911543, and NS018; a myelofibrosis inhibiting agent, including but not limited to, hedgehog inhibitors (saridegib), histone deacetylase (HDAC) inhibitors (pracinostat, panobinostat), tyrosine kinase inhibitor (lestaurtinib); a discoidin domain receptor (DDR) inhibitor, including but not limited to, those disclosed in US2009/0142345, US2011/0287011, W02013/027802, WO2013/034933, and U.S.
  • JAK inhibitor including but not limited to ruxolitinib, fedrat
  • a MMP9 inhibitor including but not limited to, marimastat (BB-2516), cipemastat (Ro 32-3555), and those described in WO2012/027721
  • a LOXL inhibitor including but not limited to the antibodies described in W02009/017833
  • a L0XL2 inhibitor including but not limited to the antibodies described in W02009/017833, W02009/035791 and WO/2011/097513
  • an ASK1 inhibitor including but not limited to, those described in W02011/008709 and WO/2013/112741
  • a PI3K6 inhibitor described in U.S. Pat. No. 7,932,260, U.S.
  • a PI3K inhibitor including but not limited to, GSK2636771, BAY 10824391, TGX221
  • a PI3Ka inhibitor including but not limited to, Buparlisib, BAY 80-6946, BYL719, PX-866, RG7604, MLN1117, WX-037, AEZS-129, PA799
  • a PI3Ky inhibitor including but not limited to, ZSTK474, AS252424, LY294002, TG100115
  • a pan PI3K inhibitor including but not limited to, LY294002, BEZ235, XL147 (SAR245408)
  • JAK inhibitors include, but not limited to, Decernotinib (or VX-509), GLPG0634, or GLPG0788
  • a MDM2 inhibitors include, but not limited to RG7112 (RO5045337), idasanutlin (RG7388), AMG-232 (KRT-232), APG-115, BI-907828, CGM097, siremadlin (HDM201), and milademetan (DS-3032b)
  • telomerase inhibitors include, but not limited to imetelstat, or a pharmaceutically acceptable salt thereof.
  • the one or more therapeutic agent is selected from the following: interferon alpha ligand modulators, such as interferon alpha-2b, interferon alpha-2a biosimilar (Biogenomics), ropeginterferon alfa-2b (AOP-2014, P-1101, PEG IFN alpha-2b), Multiferon (Alfanative, Viragen), interferon alpha lb, Roferon-A (Canferon, Ro-25-3036), interferon alfa-2a follow-on biologic (Biosidus) (Inmutag, Inter 2A), interferon alfa-2b follow-on biologic (Biosidus— Bioferon, Citopheron, Ganapar, Beijing Kawin Technology— Kaferon), Alfaferone, pegylated interferon alpha-lb, peginterferon alfa-2b follow-on biologic (Amega), recombinant human interferon alpha-lb, recombinant human interferon alpha-2a, recombin
  • Zi is OR', phenyl, naphthyl, or C5-C10 membered heterocycle, wherein the phenyl, naphthyl, or C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', - S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10- membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycl
  • Z 2 is phenyl, naphthyl, or C5-C10 membered heterocycle, wherein the phenyl, naphthyl, or C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -OR'OR", -O(CH 2 ) 2 NR'R", -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered heterocycle, and wherein the -CI-C
  • Ri is H, -C1-C6 alkyl or -C3-C7 cycloalkyl, wherein the -C1-C6 alkyl or -C3-C7 cycloalkyl is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", or - S(O) 2 R';
  • R is -C1-C6 alkyl, wherein the -C1-C6 alkyl is optionally substituted with one or more of the following: H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", or -S(O) 2 R';
  • X is a bond or -OCH(R4), wherein R4 is H, -C1-C6 alkyl or -C3-C7 cycloalkyl; R 2 and R3 are independently H, -C1-C6 alkyl, -C3-C7 cycloalkyl, aryl, C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3
  • R' or R" is independently -H or -CI-C6 alkyl, or R' and R" together, optionally attaching to N or O atom, form a 4- to 8-membered cyclic structure.
  • Zi is an optionally substituted C5-C10 membered heterocycle. In certain non-limiting implementations, Zi is an optionally substituted pyridine, pyrimidine, pyrazole, imidazole, oxazole, thiazole, indazole or tetrazole. In further non-limiting implementations, Zi is an optionally substituted pyridine or pyrazole.
  • the C5-C10 membered heterocycle, pyridine, pyrimidine, pyrazole, imidazole, oxazole, thiazole, indazole or tetrazole is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycl
  • Zi is an unsubstituted C5-C10 membered heterocycle.
  • Zi is an unsubstituted pyridine, pyrimidine, pyrazole, imidazole, oxazole, thiazole, indazole or tetrazole.
  • Zi is an unsubstituted pyridine or pyrazole.
  • Zi is an unsubstituted .
  • Zi is -
  • Z2 is an optionally substituted C5-C10 membered heterocycle.
  • Z2 is an optionally substituted phenyl, pyridine, or pyrazole.
  • the C5-C10 membered heterocycle, phenyl, pyridine, or pyrazole is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered
  • Z2 is a phenyl, pyridine or pyrazole optionally substituted with halo, -OR', -Cl- C6 alkyl, -OR'OR", or -O(CH2hNR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R", and wherein the R' or R" are independently -H, methyl or ethyl.
  • Z2 is phenyl substituted with halo, -OR', -CI-C6 alkyl, - OR'OR", or -O(CH2)2NR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R", and wherein the R' or R" are independently -H, methyl or ethyl.
  • Z2 is an unsubstituted C5-C10 membered heterocycle. In certain non-limiting implementations, Z2 is an unsubstituted phenyl, pyridine, or pyrazole. In further non-limiting implementations, Z 2 is selected from the group consisting of:
  • Ri is a H, unsubstituted methyl, methoxyethyl or dimethylaminoethyl. In some non-limiting implementations, Ri is H or In some non-limiting embodiments, R is a methyl. In other non-limiting embodiments, R is hydroxymethyl and the compound has a structure of Formula (III):
  • R is a hydroxymethyl with 5 configuration or a methyl with R configuration.
  • X is a bond.
  • X is -OCHfRzi), and the compound has a structure of
  • R2 is H
  • R3 is H, -C1-C6 alkyl, -C3-C7 cycloalkyl
  • CIO membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle, wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN,
  • R 2 is H
  • R3 is -C3-C7 cycloalkyl or -C3-C7 cycloalkyl methyl, wherein the -C3-C7 cycloalkyl or -C3-C7 cycloalkyl methyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membere
  • R 2 is H
  • R3 is O cycloalkyl selected from the group consisting of: cyclohexyl, cyclopentyl, cyclobutyl, cyclopropyl, cyclohexyl methyl, cyclopentyl methyl, cyclobutyl methyl, cyclopropyl methyl; an aryl selected from the group consisting of: phenyl and benzyl; or a C5-C10 membered heterocycle selected from the group consisting of: pyrrolidi ne-3-yl, piperidine-4-yl, (pyrrolidine-3-yl)methyl, (piperidine-4-yl)methyl, wherein the cyclohexyl, cyclopentyl, cyclobutyl, cyclopropyl, cyclohexyl methyl, cyclopentyl methyl, cyclobutyl methyl cyclopropyl methyl pheny
  • R 2 and R3 are independently H, -C3-C7 cycloalkyl, or -C3-C7 cycloalkyl methyl; wherein the -C3-C7 cycloalkyl or -C3-C7 cycloalkyl methyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 11
  • R2 and R3 are independently H, cyclohexyl, cyclopentyl, cyclobutyl, cyclopropyl, cyclohexyl methyl, cyclopentyl methyl, cyclobutyl methyl or cyclopropyl methyl; and R2 and R3 are independently optionally substituted with H, halo, -OH, - CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cyclo
  • R2 and R3 are independently H, phenyl, or benzyl; wherein the phenyl or benzyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 11-membered heterocycle; with the proviso that R2 and R3 are not both H.
  • Ri, R2, or both are H.
  • Zi is a pyridine or pyrazole
  • Z 2 is an unsubstituted phenyl, pyridine, or pyrazole, or a phenyl substituted with halo, -OR', -CI-C6 alkyl, -OR'OR", or - O(CH 2 ) 2 NR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R"
  • Ri is H, unsubstituted methyl, or dimethylamine
  • X is a bond
  • R2 is H
  • the R' or R" is independently -H, methyl or ethyl.
  • Zi is a pyridine, optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl; Z2 is a C5-C10 membered heterocycle, optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R",
  • Zi is -pyridine, -pyrimidine, -pyrazole, -imidazole, -oxazole, -thiazole, -indazole, or - tetrazole, wherein the -pyridine, -pyrimidine, -pyrazole, -imidazole, -oxazole, -thiazole, -indazole, or -tetrazole is unsubstituted or substituted with one or more of the following: -halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', - S(O) 2 NR'R", -S(O) 2 R', -guanidino, -nitro,
  • Ri is -H, -Ci-Ce alkyl, or -C3-C7 cycloalkyl, wherein the -Ci-Ce alkyl or -C3-C7 cycloalkyl is unsubstituted or substituted with one or more of the following: -halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", and -S(O) 2 R';
  • R 2 and R3 are independently -H, -Ci-Ce alkyl, -C3-C7 cycloalkyl, -aryl, a heterocycle alkyl, aryl, -C3-C7 cycloalkyl, and 3- to 11-membered heterocycle, wherein the -Ci-Ce alkyl, aryl, - C3-C7 cycloalkyl, or 3- to 11-membered heterocycle is unsubstituted or substituted with one or more of the following: -halo, -CNR'R", -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -OR'OR", -S(O)2NR'R",
  • R' and R" are independently -H or -Ci-Ce alkyl, or R' and R" together, optionally attached to N or O atom, form a 4- to 8- membered cyclic structure;
  • R 7 is -H, -halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O) 2 R', -S(O) 2 NR'R", -S(O) 2 R', -Ci-C 6 alkyl, -C3-C7 cycloalkyl, -aryl, or a heterocycle comprising 5 to 10 carbons, wherein the -Ci-Ce alkyl, -C3-C7 cycloalkyl, -aryl, or heterocycle comprising 5 to 10 carbons is unsubstituted or substituted with - -halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)
  • At least one compound for treating a myeloproliferative disorder or neoplasm is selected from the non-limiting group consisting of:
  • example compounds with a chiral center represent a racemic mixture of the corresponding R and S enantiomers, and all racemates and isolated enantiomers are within the scope of the invention. Additional non-limiting examples are:
  • At least one compound is selected from the group consisting of: Example 1
  • the following assay protocol is for measuring the phosphorylation of a peptide substrate (FAM-KKLRRTLSVA-OH wherein FAM is carboxyfluorescein).
  • the peptide is >98% purity by Capillary Electrophoresis.
  • the peptide is phosphorylated by the protein kinase ROCK or AURK.
  • the ROCK or AURK enzyme, substrate, and cofactors are combined in a well of a microtiter plate and incubated for 3 hours at 25° C. At the end of the incubation, the reaction is quenched by the addition of an EDTA-containing buffer.
  • the substrate and product are separated and quantified electrophoretically using the microfluidic-based LABCHIP® 3000 Drug Discovery System from Caliper Life Sciences (Hopkinton, Mass.).
  • Substrate and product peptides present in each sample are separated electrophoretically using the LABCHIP® 3000 capillary electrophoresis instrument. As substrate and product peptides are separated two peaks of fluorescence are observed. Change in the relative fluorescence intensity of the substrate and product peaks is the parameter measured reflecting enzyme activity. Capillary electropherograms (RDA acquisition files) are analyzed using HTS Well Analyzer software (Caliper Life Sciences, Hopkinton, Mass.). The kinase activity in each sample is determined as the product to sum ratio (PSR): P/(S+P), where P is the peak height of the product peptide and S is the peak height of the substrate peptide.
  • PSR product to sum ratio
  • the IC50 values of inhibitors are determined by fitting the inhibition curves (Pin/i versus inhibitor concentration) by 4 parameter sigmoidal dose-response model using XLfit 4 software (IBDS). This assay can be used to test the activity of each of the exemplary compounds identified in Table 2.
  • PBMC Human PBMC were plated at 5.4 5 in 6 well plates and treated with DMSO, KD025 (lOmM) or CMPD I D#10 (500,200 and lOOnM) for 1 hr. Subsequently cells were treated with IL2 10 6 U/ml and cell lysates were collected at one hour post IL-2 stimulation. Lysates were prepared in radioimmunoprecipitation assay buffer (RIPA buffer) containing protease and phosphatase inhibitors.
  • RIPA buffer radioimmunoprecipitation assay buffer
  • MLC2 Myosin light chain
  • pMLC2 phosphorylated
  • Human Hel 92.1.7 and UKE1 cells were plated at 5.0xl0 5 cells per well of a 6 well dish. 24 hours after plating, cells were treated with DMSO or CMPD #97 (1.0 mm or 0.1 mM) in duplicate. Cells were collected at 24 and 48 hours and lysates prepared in IX lithium dodecyl sulfate (LDS) buffer (Life Technologies, Carlsbad, CA) supplemented with protease- and phosphatase inhibitors (Roche). Equal volumes of protein were run on 4-12 % gels and transferred to PVDF membranes.
  • LDS lithium dodecyl sulfate
  • Blots were developed with horseradish peroxidase conjugated secondary antibodies and imaged on a MP3 imager (Bio-Rad).
  • Ratio of phosphorylated MLC2 to MLC2 was calculated and expressed as percentage of the control set to 100%
  • CMPD #97 significantly decreased phosphorylation of MLC2 (pMLC2) at 1.0 mM and 0.1 mM concentrations, in both human cell lines confirming an on-target inhibition of ROCK.
  • CFU Colony Formation Unit
  • CMPD #97 DMSO solution with MethoCult H4230 media (StemCell, #04230)
  • 16,500 cells in 0.3 mL cell culture media were added into 3.0 mL MethoCult media in a 5 mL tube.
  • Cells were resuspended into the MethoCult media by gentle agitation. Tubes were kept standing at room temperature for 15 minutes.
  • Cells were placed by a syringe with a blunt end needle into 6-well plates at 5,000 cells/1.0 mL/well. Plates were kept in a cell culture incubator for 7 days.
  • colony size in control group (DMSO only) reached 50-100 pm, 5-6 images were randomly taken under 4x magnification for each condition, and colony number count by a blinded observer.
  • CMPD I D#97 in human HEL-92.1.7 cells The ICso for CMPD I D#97 in human HEL-92.1.7 cells was 25.21 nM. CMPD I D#97 was serially diluted 3:1 from 10 pM (#12) to 0.17 nM (#2). Condition 1 (#1) was DMSO control. See Figure 7. The IC50 for CMPD ID#97 in human UKE-1 cells was 54.62 nM. CMPD I D#97 was serially diluted 3:1 from 10 pM (#9) to 4.57 nM (#2). Condition 1 (#1) was DMSO control. See Figure 8.
  • the cell lines HEL 92.1.7 and UKE-1 were synchronized with 2mM Thymidine (Cayman Chemical (Ann Arbor, Ml) for 18hr, washed, and rested for4hr.
  • the cells were treated with CMPD #10 and CMPD #97 for 24hr at 500nM and lpM. After treatment, the cells were harvested, washed twice in IX DPBS, and fixed in 70% ice-cold ethanol for lhr on ice. After fixation cells were washed with IX DPBS and let rehydrate for 15 min.
  • the samples were analyzed using the MACSQuant Analyzer 16 (Miltenyi Biotec (Bergisch Gladback, Germany)) and a total of 20,000 single cell events were recorded per sample.
  • FlowJo vlO BD Biosciences (Franklin Lakes, NJ) was used to analyze the data. Doublets were excluded from the analysis by plotting the side scatter width vs. side scatter area.
  • the cell cycle distribution was calculated using the Watson Pragmatic Model. Each condition had two biological replicates that were split into three technical replicates for staining and analysis. See Figures 9, 10, 11.
  • CMPD ID#97 was added to MethoCult Media (standardized semi-solid methylcellulose media with appropriate cytokines for colony assays) at varying concentrations (0 mM (control), 0.06 mM, 0.19 mM, and 0.56 mM).
  • CMPD ID#97- containing MethoCult Media 3,000 cells per mL
  • the CFU assay was allowed to grow for 12 days in a humidified incubator at 37°C, 5% CO 2 .
  • colonies were identified and counted for each CFU type (burst forming colonies of erythrocytes (BFU), granulocytes and macrophages (GM), and granulocytes, erythrocytes, macrophages, and megakaryocytes (GEMM).
  • BFU erythrocytes
  • GM granulocytes and macrophages
  • GEMM megakaryocytes
  • mice Six-week-old female CIEA NOG mice (NOD.Cg-Prkdcscid H2rgtmlSug/JicTac), received from Taconic, were inoculated with human HEL-92.1.7 (7.5xl0 5 cells/mouse) intravenously using a saline vehicle. Mice were randomized into vehicle and treatment groups by body weights five days following tumor cell inoculation and treatment was initiated. Experimental test article CMPD #97 (in 10% DMA: 90% PEG400 vehicle) was administered orally daily at 5 and 25 mg/kg. Ruxolitinib (in 10% DMA: 90% PEG400 vehicle) was also administered orally daily at 45 mg/kg alone or in combination with CMPD #97. Body weights were measured thrice weekly. Study endpoint was time to peri-morbidity to assess tumor growth delay of treated groups compared to vehicle control. See Figure 14.

Landscapes

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

Abstract

Compositions useful for treating a subject suffering from a myeloproliferative disorder or disease, or from a disease or condition associated with myeloproliferative neoplasms, and methods of such treatment using the compositions, wherein the compositions include an effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.

Description

COMPOSITIONS AND METHODS TO TREAT MYELOPROLIFERATIVE DISORDERS AND MYELOPROLIFERATIVE NEOPLASMS
CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority in U.S. Provisional Patent Application No. 63/399,616, filed on August 19, 2022, the content of which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
The present application provides the compositions and methods for treating myeloproliferative disorders or neoplasms. The application also provides the methods for preparation of the compositions, the article of manufacture, and the kit thereof.
BACKGROUND ART
Myeloproliferative disorders or neoplasms are caused by genetic defects in the hematopoietic stem cells, resulting in clonal myeloproliferation, bone marrow fibrosis, and abnormal cytokine expression (Tefferi et al Cancer Res 2022;82:749-63). MPN may be classified into four subtypes: chronic myelogenous leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). Treatments of myeloproliferative disorders involve allogeneic stem cell transplant. The transplant procedure is preceded by myeloablative chemotherapy, can lead to severe treatment-related consequences such as graft- versus-host disease, and is limited by performance status, age, and donor restrictions.
In 2005, a mutation JAK2V617F in Janus kinase 2 or JAK2, a member of the JAK family of kinases was identified (Baxter et aL, Lancet 365:1054-61, 2005; James et al., Nature 434:1144-8, 2005; Kralovics et al., N. Engl. J. Med. 352:1779-90, 2005; Levine et al., Cancer Cell 7:387-97; 2005). The mutation constitutively activates JAK2 and JAK-STAT signaling, resulting in unrestrained cellular proliferation characteristics of myeloproliferative disorders. It is found in the subtypes of PV, ET, and PMF. About 99% of polycythemia vera patients and about 50-60% of essential thrombocytopenia patients and idiopathic myelofibrosis patients have the mutation jAK2V517F (Vainchenker et al.. Blood 118:1723-35, 2011). Several JAK inhibitors have been developed for treating myeloproliferative neoplasms, including ruxolitinib (INCB018424) for treating primary myelofibrosis, fedratinib (SAR302503, TG101348) for treating myelofibrosis, and XL019, SB1518 and AZD1480 for treating post-PV/ET myelofibrosis (Sonbol, Ther. Adv. Hematol. 4: 15-35, 2013). Patients treated with JAK inhibitors exhibit clinical improvement of reduced splenomegaly and/or constitutional symptoms. However, certain patients' anemia and thrombocytopenia conditions are aggravated. CYT387 (momelotinib) or N-(cyanomethyl)-4-(2-(4-morpholinophenylamino) pyrimidin-4-yl)benzamide is a different class of JAK inhibitor that provide additional benefits in improving anemia and/or spleen response. It is currently in clinical trials for treating primary myelofibrosis, polycythemia vera (PV), essential thrombocythemia (ET), and post-PV/ET.
It is shown that patients who have received chronic ruxolitinib treatment commonly develop disease persistence as shown by the gradual return of splenomegaly and/or constitutional symptoms, the lack of hematologic or molecular remissions, or the loss of clinical improvement (Gotlib, Hematologist, November 2012:11).
Accordingly, there is a need of effective treatment of myeloproliferative disorders including progressive or relapsed disease.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 depicts gel blot results demonstrating effects on phosphorylated STAT3 in IL-2 activated human peripheral blood mononuclear cells (PBMC).
Fig. 2 plots the dose dependent inhibitory effect of compound ID#97 on STAT3 phosphorylation in PBMC, compared to KD025 (belumosudil).
Fig. 3 show a Western blot of total MLC2 and pMLC2 levels in human HEL 92.1.7 cells after treatment with compound I D#97.
Fig. 4 is a bar chart quantifying the effect of compound ID#97 on MLC phosphorylation in human HEL 92.1.7 cells.
Fig. 5 is a Western blot of total MLC2 and pMLC2 levels in human UKE-1 Cells after treatment with compound I D#97.
Fig. 6 is a bar chart quantifying the effect of compound ID#97 on MLC phosphorylation in human UKE-1 cells. Fig. 7 depicts CFU assay in human HEL-92.1.7 cells with serial dilution of compound ID#97.
Fig. 8 depicts CFU assay in human UKE-1 cells with serial dilution of compound ID#97.
Fig. 9 is a cell cycle analysis in human HEL-92.1.7 cells.
Fig. 10 is a cell cycle analysis in human UKE-1 cells.
Fig. 11 shows cell cycle distributions for HEL-92.1.7 cells and UKE-1 cells, after treatment with compound #10 or compound #97.
Fig. 12 is a bar graph depicting colony forming unit assay results.
Fig. 13 is a summary of the colony forming unit assay results, listing mean CFU +/- SEM.
Fig. 14 plots in vivo survival rates after i.v. treatment with compound #97, with or without Ruxolitinib, on HEL-92.1.7 cells.
DISCLOSURE
An important large family of enzymes is the protein kinase enzyme family. Currently, there are about 500 different known protein kinases. Protein kinases serve to catalyze the phosphorylation of an amino acid side chain in various proteins by the transfer of the y-phosphate of the ATP-Mg2+ complex to said amino acid side chain. These enzymes control the majority of the signaling processes inside cells, thereby governing cell function, growth, differentiation, and destruction (apoptosis) through reversible phosphorylation of the hydroxyl groups of serine, threonine, and tyrosine residues in proteins. Studies have shown that protein kinases are key regulators of many cell functions, including signal transduction, transcriptional regulation, cell motility, and cell division. Several oncogenes have also been shown to encode protein kinases, suggesting that kinases play a role in oncogenesis. These processes are highly regulated, often by complex intermeshed pathways where each kinase will itself be regulated by one or more kinases. Consequently, aberrant, or inappropriate protein kinase activity can contribute to the rise of disease states associated with such aberrant kinase activity. Due to their physiological relevance, variety and ubiquitousness, protein kinases have become one of the most important and widely studied family of enzymes in biochemical and medical research.
The protein kinase family of enzymes is typically classified into two main subfamilies: Protein Tyrosine Kinases and Protein Serine/Threonine Kinases, based on the amino acid residue they phosphorylate. The serine/threonine kinases (PSTK), includes cyclic AMP- and cyclic GMP- dependent protein kinases, calcium- and phospholipid-dependent protein kinase, calcium- and calmodulin-dependent protein kinases, casein kinases, cell division cycle protein kinases and others. These kinases are usually cytoplasmic or associated with the particulate fractions of cells, possibly by anchoring proteins. Aberrant protein serine/threonine kinase activity has been implicated or is suspected in several pathologies such as rheumatoid arthritis, psoriasis, septic shock, bone loss, many cancers, and other proliferative diseases. Accordingly, serine/threonine kinases and the signal transduction pathways which they are part of are important targets for drug design. The tyrosine kinases phosphorylate tyrosine residues. Tyrosine kinases play an equally important role in cell regulation. These kinases include several receptors for molecules such as growth factors and hormones, including epidermal growth factor receptor, insulin receptor, platelet derived growth factor receptor and others. Studies have indicated that many tyrosine kinases are transmembrane proteins with their receptor domains located on the outside of the cell and their kinase 5 domains on the inside. Much work is also under progress to identify modulators of tyrosine kinases as well.
A major signal transduction system utilized by cells is the RhoA-signaling pathways. RhoA is a small GTP binding protein that can be activated by several extracellular stimuli such as growth factor, hormones, mechanic stress, osmotic change as well as high concentration of metabolite like glucose. RhoA activation involves GTP binding, conformation alteration, post-translational modification (geranylgeranylation and famesylation) and activation of its intrinsic GTPase activity. Activated RhoA can interact with several effector proteins including ROCKs and transmit signals into cellular cytoplasm and nucleus.
ROCK1 and 2 (ROCK) constitute a family of kinases that can be activated by RhoA-GTP complex via physical association. Activated ROCKs phosphorylate several substrates and play important roles in pivotal cellular functions. The substrates for ROCKs include myosin binding subunit of myosin light chain phosphatase (MBS, also named MYPT1), adducin, myosin light chain (MLC), LIM kinase as well as transcription factor FHL. The phosphorylation of theses substrates modulates the biological activity of the proteins and thus provide a means to alter cell's response to external stimuli. One well documented example is the participation of ROCK in smooth muscle contraction. Upon stimulation by phenylephrine, smooth muscle from blood vessels contracts. Studies have shown that phenylephrine stimulates alpha-adrenergic receptors and leads to the activation of RhoA. Activated RhoA in turn stimulates kinase activity of ROCK1 and which in turn phosphorylates MBS. Such phosphorylation inhibits the enzyme activity of myosin light chain phosphatase and increases the phosphorylation of myosin light chain itself by a calciumdependent myosin light chain kinase (MLCK) and consequently increases the contractility of myosin-actin bundle, leading to smooth muscle contraction.
This phenomenon is also sometimes called calcium sensitization. In addition to smooth muscle contraction, ROCKs have also been shown to be involved in cellular functions including apoptosis, cell migration, transcriptional activation, fibrosis, cytokinesis, inflammation, and cell proliferation. Moreover, in neurons ROCK plays a critical role in the inhibition of axonal growth by myelin-associated inhibitory factors such as i myelin-associated glycoprotein (MAG). ROCK- activity also mediates the collapse of growth cones in developing neurons.
Both processes are thought to be mediated by ROCK-induced phosphorylation of substrates such as LIM kinase and myosin light chain phosphatase, resulting in increased contractility of the neuronal actin-myosin system. Inhibitors of ROCKs have been suggested for use in the treatments of a variety of diseases. They include cardiovascular diseases such as hypertension, chronic and congestive heart failure, cardiac hypertrophy, restenosis, chronic renal failure, and atherosclerosis. In addition, because of its muscle relaxing properties, it is also suitable for asthma, male erectile dysfunctions, female sexual dysfunction, and over-active bladder syndrome. ROCK inhibitors have been shown to possess anti-inflammatory properties. Thus, they can be used as treatment for neuroinflammatory diseases such as stroke, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and inflammatory pain, as well as other inflammatory diseases such as rheumatoid arthritis, irritable bowel syndrome, inflammatory bowel disease. In addition, based on their neurite outgrowth inducing effects, ROCK inhibitors could be useful drugs for neuronal regeneration, inducing new axonal growth and axonal rewiring across lesions within the CNS. ROCK inhibitors are therefore likely to be useful for regenerative (recovery) treatment of CNS disorders such as spinal cord injury, acute neuronal injury (stroke, traumatic brain injury), Parkinson's disease, Alzheimer's disease, and other neurodegenerative disorders. Since ROCK inhibitors reduce cell proliferation and cell migration, they could be useful in treating cancer and tumor metastasis. Furthermore, there is evidence suggesting that ROCK inhibitors suppress cytoskeletal rearrangement upon virus invasion, thus they also have potential therapeutic value in antiviral and anti-bacterial applications. ROCK inhibitors may also be useful for the treatment of insulin resistance and diabetes.
The aurora family of serine/threonine protein kinases is essential for cell proliferation. The human aurora kinase family consists of three highly homologous kinases (A or "2", B or "1" and C or "3"). During normal cell proliferation, these proteins are involved in chromosome segregation, mitotic spindle function, and cytokinesis. Aurora kinase expression is low in resting cells and peaks during the G2 and mitosis phases of the cell cycle. Several proposed mammalian substrates for Aurora kinases that are important for cell division include histone H3, TPX2, myosin II regulatory light chain, CENP-A, and protein phosphatase 1.
Since the elucidation of their key role in mitotic progression and cell division, Aurora kinases have been closely linked to tumorigenesis. For example, Aurora kinase gene amplification and overexpression has been reported in many cancers. A coding single nucleotide polymorphism (SNP) has been identified that is significantly more frequent in advanced gastric cancer relative to early-stage gastric cancer, and this SNP correlates with elevated kinase activity. Overexpression of Aurora A induces centrosome amplification, aneuploidy, and transformation in rodent fibroblasts. This oncogenic activity is likely due to the generation of chromosome instability. Indeed, there is a strong correlation between Aurora A overexpression and chromosome aneuploidy in breast and gastric cancer.
Aurora B expression is elevated in cell lines derived from tumors of the colon, breast, lung, melanoma, kidney, ovary, pancreas, CNS, gastric tract and leukemias. In prostate cancer, increased nuclear expression of Aurora B was observed in high Gleason grade anaplastic prostate cancer tissues relative to low and intermediate grades, and Aurora B expression was accompanied by the phosphorylation of the histone H3 substrate. Because Aurora kinase inhibition in tumor cells can result in mitotic arrest and apoptosis, these kinases are important targets for cancer therapy. Given the central role of mitosis in the progression of virtually all malignancies, inhibitors of the Aurora kinases therefore are expected to have the potential to block growth of cancers or tumors and have application across a broad range of human cancers.
ROCK1 and ROCK2 expression is elevated in patients with essential thrombocythemia (ET), myelofibrosis (MF), and polycythemia vera (PV) compared to healthy individuals (Saleiro et al, Nature Communications (2022) 13:1750). ROCK kinases and aurora kinases are involved in megakaryocyte differentiation and maturation, platelet production, and proplatelet formation and this is implicated in myeloproliferative neoplasms (Yang et al, Exp Hematol. 2017 April ; 48: 32-38)
Provided herein are methods, compositions, articles of manufacture, and kits for treating a hyperproliferative disorder by using effective amounts of one, two or more therapeutic agents including a ROCK inhibitor, a Janus kinase (JAK) inhibitor, or the combination thereof. The methods described herein provide a treatment for a myeloproliferative disorder, comprising administering to a patient a therapeutic effective amount of JAK inhibitor and a therapeutic effective amount of a ROCK inhibitor.
In one aspect of the application, the JAK inhibitor is selected from the group consisting of ruxolitinib, fedratinib, tofacitinib, baricitinib, lestaurtinib, pacritinib, XL019, AZD1480, INCB039110, LY2784544, BMS911543, NS018, or N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide; or pharmaceutically acceptable salts thereof. In one embodiment, the JAK2 inhibitor is ruxolitinib. In other embodiment, the JAK inhibitor is a JAK2 inhibitor N-(cyanomethyl)-4-[2-(4-morpholinoanilino)pyrimidin-4-yl]benzamide or a pharmaceutically acceptable salt thereof. In some aspect, the JAK inhibitors are selected from Decernotinib (or VX-509), GLPG0634, or GLPG0788, or a pharmaceutically acceptable salt thereof.
The method of treating myeloproliferative diseases further comprises one or more therapeutic agents selected from a spleen tyrosine kinase (SYK) inhibitor, a Bruton's tyrosine kinase (BTK) inhibitor, a bromodomain-containing protein (BRD) inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anticancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. Additional methods include one or more therapeutic agent selected from a PI3K (including PI 3Ky, PI3K6, PI3KI 3, and PI3Ka inhibitor, a JAK (including JAK1 and JAK2) inhibitor, a SYK inhibitor, a BTK inhibitor, a BRD (including BRD4 inhibitor), a, BCL2 inhibitor, a LOXL (including LOXL1, LOXL2, LOXL3, LOXL4, or LOXLS) inhibitor, a MMP (including MMP2 and MMP9) inhibitor, a A2B inhibitor, an IDH (including IDH1) inhibitor, an ASK (including ASK1) inhibitor, a TPL2 inhibitor, a DDR (including DDR1 and DDR2) inhibitor, an HDAC inhibitor, a PKC inhibitor, or any combination thereof. In some aspect, one or more therapeutic agents are selected from an Abl inhibitor, an ACK inhibitor, an A2B inhibitor, an ASK inhibitor, an Aurora kinase inhibitor, a BTK inhibitor, a BRD inhibitor, a c-Kit inhibitor, a c-Met inhibitor, a CAK inhibitor, a CaMK inhibitor, a CDK inhibitor, a CK inhibitor, a DDR inhibitor, an EGFR inhibitor, a FAK inhibitor, a Flt-3 inhibitor, a FYN inhibitor, a GSK inhibitor, a HCK inhibitor, a HDAC inhibitor, an IKK inhibitor, an IDH inhibitor, an IKK inhibitor, a KDR inhibitor, a LCK inhibitor, a LOX inhibitor, a LOXL inhibitor, a LYN inhibitor, a MMP inhibitor, a MEK inhibitor, a MAPK inhibitor, a MDM2 inhibitor, a NEK9 inhibitor, a NPM-ALK inhibitor, a p38 kinase inhibitor, a PDGF inhibitor, a PK inhibitor, a PLK inhibitor, a PK inhibitor, a PYK inhibitor, a SYK inhibitor, a TPL2 inhibitor, a STK inhibitor, a STAT inhibitor, a SRC inhibitor, a TBK inhibitor, a TIE inhibitor, a TK inhibitor, a telomerase inhibitor, a VEGF inhibitor, a YES inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof.
The myeloproliferative disorder is selected from the group consisting of polycythemia vera (PV), primary myelofibrosis (PMF), thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), chronic myelogenous leukemia (CML), systemic mastocystosis (SM), chronic neutrophilic leukemia (CNL), myelodysplastic syndrome (MDS) and systemic mast cell disease (SMCD). In one aspect, the myeloproliferative disorder is myelofibrosis (MF).
In other aspect of the application, a treatment is provided for patients having myeloproliferative disorder selected from the group consisting of polycythemia vera (PV), primary myelofibrosis (PMF), or essential thrombocythemia (ET). The patient has received prior treatment and/or develops disease persistence to treatment of myeloproliferative disorder or has not previously been treated for myeloproliferative disorder. In additional aspect of the application, a treatment is provided for patients having diseases selected from diffuse large B- cell lymphoma.
In some other aspect, a method for decreasing cell viability, decreasing proliferation, or increasing apoptosis is provided. Such methods comprise contacting cells with an effective amount of JAK inhibitor and an effective amount of PI3K inhibitor. The JAK inhibitor is selected from the group consisting of ruxolitinib, fedratinib, tofacitinib, baricitinib, lestaurtinib, pacritinib, XL019, AZD1480, INCB039110, LY2784544, BMS911543, NS018, or N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide; or pharmaceutically acceptable salts thereof. The method uses cells that are isolated from a subject having myeloproliferative disorder selected from the group consisting of polycythemia vera (PV), primary myelofibrosis (PMF), thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), chronic myelogenous leukemia (CML), systemic mastocystosis (SM), chronic neutrophilic leukemia (CNL), myelodysplastic syndrome (MDS) and systemic mast cell disease (SMCD). Also, the method uses cells that are isolated from a subject having diffuse large B-cell lymphoma (DLBCL).
In some aspects, a pharmaceutical composition comprising a therapeutically effective amount of JAK inhibitor, a therapeutically effective amount of ROCK inhibitor, and a pharmaceutically acceptable excipient is provided.
In certain aspects, a kit comprising a pharmaceutical composition and a label is provided. The kit contains the pharmaceutical composition that comprises a therapeutically effective amount of JAK inhibitor, a therapeutically effective amount of ROCK inhibitor, and a pharmaceutically acceptable excipient.
In one aspect the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a myeloproliferative disorder. In one aspect the application provides a JAK2 inhibitor N-(cyanomethyl)-4-[2-(4-morpholinoanilino)pyrimidin-4-yl]benzamide; or a pharmaceutically acceptable salt thereof, which is administered at a dose between 50 to 350 mg; or between 100 to 200 mg.
In one aspect the method of treating myeloproliferative diseases further comprises one or more therapeutic agents selected from a SYK inhibitor, a BTK inhibitor, a BRD inhibitor, a MDM2 inhibitor, a telomerase inhibitor, a BCL2 inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. In one aspect, the administration of the JAK inhibitor is prior to the administration of the ROCK inhibitor. In another aspect, the administration of the JAK inhibitor is concurrent to the administration of the ROCK inhibitor. In another aspect, the administration of the JAK inhibitor is after the administration of the ROCK inhibitor.
In certain aspects, the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a hyperproliferative disorder. In some aspect, the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a hyperproliferative disorder. In additional aspect, the application provides a ROCK inhibitor for use in a method for treating a hyperproliferative disorder. In other aspect, the application provides a ROCK inhibitor for use in a method fortreating a hyperproliferative disorder. In one aspect, the hyperproliferative disorder is myeloproliferative disorder. In other aspect, the hyperproliferative disorder is cancer. In additional aspect, the application provides a ROCK inhibitor for use in treating hyperproliferative disorders or neoplasms, wherein the ROCK inhibitor is administered simultaneously, separately, or sequentially with a ROCK inhibitor.
In one aspect, the method of treating hyperproliferative diseases comprising administering a therapeutically effective amount of an Abl inhibitor, an ACK inhibitor, an A2B inhibitor, an ASK inhibitor, an Aurora kinase inhibitor, a BTK inhibitor, a BRD inhibitor, a BCL2 inhibitor, a c-Kit inhibitor, a c-Met inhibitor, a CAK inhibitor, a CaMK inhibitor, a CDK inhibitor, a CK inhibitor, a DDR inhibitor, an EGFR inhibitor, a FAK inhibitor, a Flt-3 inhibitor, a FYN inhibitor, a GSK inhibitor, a HCK inhibitor, a HDAC inhibitor, an IKK inhibitor, an IDH inhibitor, an IKK inhibitor, a JAK inhibitor, a KDR inhibitor, a LCK inhibitor, a LOX inhibitor, a LOXL inhibitor, a LYN inhibitor, a MMP inhibitor, a MEK inhibitor, a MARK inhibitor, a MDM2 inhibitor, a NEK9 inhibitor, a NPM-ALK inhibitor, a p38 kinase inhibitor, a PDGF inhibitor, a PIS kinase (PI3K), a PK inhibitor, a PLK inhibitor, a PK inhibitor, a PYK inhibitor, a SYK inhibitor, a TPL2 inhibitor, a STK inhibitor, a STAT inhibitor, a SRC inhibitor, a TBK inhibitor, a TIE inhibitor, a telomerase inhibitor, a TK inhibitor, a VEGF inhibitor, a YES inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti- proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. In certain aspect, the one or more therapeutic agent is selected from a ROCK inhibitor, a JAK (including JAK1 and JAK2) inhibitor, a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor, a SYK inhibitor, a BTK inhibitor, a BRD (including BRD4 inhibitor), a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti- neoplastic agent, an anti-cancer agent, an anti-proliferation agent, or any combination thereof.
In some aspects, the application provides a JAK inhibitor and a ROCK inhibitor for use in a method for treating a myeloproliferative disorder. In additional aspect, the application provides a ROCK inhibitor for use in a method for treating a myeloproliferative disorder. In other aspect, the application provides a ROCK inhibitor for use in a method for treating a myeloproliferative disorder. In other aspect, the administration of the JAK inhibitor is prior to the administration of the ROCK inhibitor. In one aspect the application provides a JAK2 inhibitor N-(cyanomethyl)-4-[2- (4-morpholinoanilino)pyrimidin-4-yl]benzamide; or a pharmaceutically acceptable hydrochloride salt thereof, which is administered at a dose between 100 to 300 mg. In additional aspect, the application provides a JAK inhibitor ruxolitinib, or a pharmaceutically acceptable phosphate salt thereof, which is administered at a dose between 15 to 25 mg.
DESCRIPTION OF EMBODIMENTS
The following description sets forth exemplary methods, compositions, kits, and articles of manufacture for treating myeloproliferative disorders or neoplasm. Such description exemplifies embodiments and does not limit the scope of the present disclosure.
The present application provides methods for treating hyperproliferative disorders such as cancers and myeloproliferative disorders in a subject by administering one or more therapeutic agents. The myeloproliferative disorders (MPD), also referred to as myeloproliferative neoplasms (MPN), are caused by mutations in the hematopoietic (or early myeloid progenitor) stem cells that result in excessive production of myeloid lineage cells (such as bone marrow), clonal myeloproliferation, bone marrow fibrosis, and abnormal cytokine expression. MPN includes, among others, polycythemia vera (PV), primary myelofibrosis, thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis, chronic myelogenous leukemia (CIVIL), systemic mastocystosis, chronic neutrophilic leukemia, myelodysplastic syndrome, and systemic mast cell disease. MPN patients may further develop acute myeloid leukemia (AML), which is often associated with a poor outcome. Current MPN therapies aim at providing palliative care over a long period of time.
The methods provided herein treat myeloproliferative diseases by administering one or more therapeutic agents for treating myeloproliferative diseases. In certain embodiments, the methods use or include a single therapeutic agent. In other embodiment, the methods use or include a combination of two or more therapeutic agents. In some embodiments, a method is provided for treating myeloproliferative diseases by administering a combination of therapeutic agents or small molecule inhibitors that inhibit B-cell receptor (BCR)-mediated signaling, phosphatidylinositol 3-kinase (PI3K)-mediated, Janus kinase (JAK)-mediated signaling pathways, BCL2 inhibitors, MDM2 inhibitors, telomerase inhibitors or any combination thereof.
A therapeutic agent may be a compound or a biologic molecule (such as DNA, RNA, or protein) that provide desired therapeutic effects when administered to a subject in need thereof (e.g., MPN patients). For example, the therapeutic agent is a compound that inhibits kinase that, directly or indirectly, relates to the disease mechanism or development. As used herein, enhanced therapeutic effects or variants thereof refer to additional beneficial or synergistic effects to patients that are not observed previously, including fewer and/or reduced symptoms, higher survival rate, prolonged survival time, shorter treatment duration, lower drug dosage, increased molecular and/or cellular responses, and the like.
The combination of therapeutic agents or inhibitors may target upstream or downstream components of the same pathway. Alternatively, the combination of therapeutic agents or inhibitors may target different components of dual or multiple pathways. It is hypothesized that the use of a combination of therapeutic agents or inhibitors may enhance therapeutic effects compared to the use of a single therapeutic agent or inhibitor.
Regardless of the mechanism, such effects are desired in treating hyperproliferative diseases such as cancers and MPN as the treatment is generally provided over a long period of time (i.e., chronic therapies) and drug resistance or disease persistence are commonly observed during chronic therapies. Thus, dual, or multiple inhibitions by a combination of two, three or more therapeutic agents may enhance treatment or therapeutic effects in myeloproliferative diseases.
The application also provides compositions (including pharmaceutical compositions, formulations, or unit dosages), articles of manufacture and kits comprising one or more therapeutic agents, including a ROCK inhibitor, a PI3K inhibitor (including a PI3K6 inhibitor), a spleen tyrosine kinase (SYK) inhibitor, a Janus kinase (JAK) inhibitor (including a JAK2 inhibitor), a Bruton's tyrosine kinase (BTK) inhibitor, and a bromodomain containing protein inhibitor (BRD) inhibitor (including a BRD4 inhibitor), a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor. In some embodiments, one or more therapeutic agent is selected from a PI3K (including PI3Ky, PI3K6, PI3K , PI3Ka, and/or pan-PI3K) inhibitor, a JAK (including JAK1 and/or JAK2) inhibitor, a SYK inhibitor, a BTK inhibitor, a BCL2 inhibitor, an A2B (adenosine A2B receptor) inhibitor, an ACK (activated CDC kinase, including ACK1) inhibitor, an ASK (apoptosis signal-regulating kinase, including ASK1) inhibitor, Aurora kinase, a BRD (bromodomain-containing protein, including BRD4) inhibitor, a BCL2 inhibitor, a CAK (CDK-activating kinase) inhibitor, a CaMK (calmodulindependent protein kinases) inhibitor, a CDK (cyclin-dependent kinases, including CDK1, 2, 3, 4, and/or 6) inhibitor, a CK (casein kinase, including CK1 and/or CK2) inhibitor, a DDR (discoidin domain receptor, including DDR1 and/or DDR2) inhibitor, a EGFR inhibitor, a FAK (focal adhesion kinase) inhibitor, a GSK (glycogen synthase kinase) inhibitor, a HDAC (histone deacetylase) inhibitor, an IDH (isocitrate dehydrogenase, including IDH1) inhibitor, an IKK inhibitor, a LCK (lymphocyte-specific protein tyrosine kinase) inhibitor, a LOX (lysyl oxidase) inhibitor, a LOXL (lysyl oxidase like protein, including LOXL1, LOXL2, LOXL3, LOXL4, and/or LOXL5) inhibitor, a MEK inhibitor, a matrix metalloprotease (MMP, including MMP2 and/or MMP9) inhibitor, a mitogen- activated protein kinases (MAPK) inhibitor, a MDM2 inhibitor, a PDGF (platelet-derived growth factor) inhibitor, a phosphorylase kinase (PK) inhibitor, a PLK (polo-like kinase, including PLK1, 2, 3) inhibitor, a protein kinase (PK, including protein kinase A, B, C) inhibitor, a serine/threonine kinase (STK) inhibitor, a STAT (signal transduction and transcription) inhibitor, a TBK (serine/threonine-protein kinase, including TBK1) inhibitor, a TK (tyrosine kinase) inhibitor, a TPL2 (serine/threonine kinase) inhibitor, a telomerase inhibitor, a NEK9 inhibitor, an Abl inhibitor, a p38 kinase inhibitor, a PYK inhibitor, a PYK inhibitor, a c-Kit inhibitor, a NPM-ALK inhibitor, a Flt- 3 inhibitor, a c-Met inhibitor, a KDR inhibitor, a TIE-2 inhibitor, a VEGFR inhibitor, a SRC inhibitor, a HCK inhibitor, a LYN inhibitor, a FYN inhibitor, a YES inhibitor, or any combination thereof. By way of example, the therapeutic agents include a ROCK inhibitor, or a pharmaceutically acceptable salt thereof, and a JAK2 inhibitor, or a pharmaceutically acceptable salt thereof.
As described in the present application, the administration of a ROCK inhibitor, and a JAK inhibitor, including N-(cyanomethyl)-4-(2-((4-morpholinophenyl)amino)pyrimidin-4-yl)benza- mide or ruxolitinib, to diseased cells or patients has led to unexpected enhanced therapeutic effects compared to the administration of each kinase inhibitor alone. The unexpected synergistic effects include, but are not limited to, for example, decreased cell viability, increased cell death or apoptosis, decreased inhibition or interference with ROCK signaling pathways, and/or reduction in chemokine (e.g., CCL2, CCL3, CLL4 and CLL22) production, reduced colony formation in diseased cells or patients. Also, unexpected effects may include, but are not limited to, increased inhibition or interference of JAK/STAT (including STAT3 and STAT5), decreased doses or duration of a single agent treatment. Further, the administration of both ROCK and JAK inhibitors unexpectedly restored or increased sensitivity or response of the diseased cells that had developed resistance, or the patients developed disease persistence to prior treatment.
THERAPEUTIC AGENTS
The present application provides methods, compositions, kits, and articles of manufacture thereof that use or include one or more therapeutic agents inhibiting one or more targets that relate to, directly or indirectly, cell growth, proliferation, or apoptosis for treating hyperproliferative disorders such as cancers or myeloproliferative neoplasms. The one or more therapeutic agents are compounds or molecules that target a PI3 kinase (PI3K), a spleen tyrosine kinase (SYK), a Janus kinase (JAK), a bromodomain-containing (BRD), a Bruton's tyrosine kinase (BTK), a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor, or any combination thereof, resulting in the inhibition of the target. In certain embodiments, the therapeutic agent is a ROCK inhibitor that inhibits ROCK1 and ROCK2. In some embodiments, the therapeutic agents are a ROCK inhibitor and a JAK2 inhibitor. The JAK inhibitor binds and inhibits one or more members of JAK family, including JAK1, JAK2, and/or JAK3. In one embodiment, the JAK inhibitor is N-(cyanomethyl)-4-[2-(4- morpholinoanilino)pyrimidin-4-yl]benzamide, also referred to as CYT0387 or momelotinib, that is a selective inhibitor to JAK2 and JAK1, relative to JAK3. Methods for synthesizing CYT0387 or momelotinib are previously described in U.S. Pat. No. 8,486,941. This reference is hereby incorporated herein by reference in its entirety. Additional JAK inhibitors include, but are not limited to, ruxolitinib (INCB018424), fedratinib (SAR302503, TG101348), tofacitinib, baricitinib, lestaurtinib, pacritinib (SB1518), XL019, AZD1480, INCB039110, LY2784544, BMS911543, and NS018. Other JAK inhibitors include, but not limited to, Decernotinib (or VX-509), GLPG0634, or GLPG0788, or a pharmaceutically acceptable salt thereof.
One skilled in the art understands that the compound structures may be named or identified using commonly recognized nomenclature systems and symbols. By way of example, the compound may be named or identified with common names, systematic or non-systematic names The nomenclature systems and symbols that are commonly recognized in the art of chemistry include, for example, ChemBioDraw Ultra 12.0, Chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (IUPAC).
In some embodiments, the one or more therapeutic agents include inhibitors that are being used and/or developed to treat various hyperproliferative disorders such as cancer or myeloproliferative neoplasms. Exemplified therapeutic agents include compounds or molecules inhibiting pathways related to BCR, BCL2. PI3K, MDM2, telomerase, SYK, and JAK, such as the agents inhibiting the RAS/RAFMEK/ERK pathway, the PI3K/PTEN/AKT/mTOR pathway, and the JAK-STAT pathway Inhibitors of mTOR include temsirolimus, everolimus, ridaforolimus (or deforolimus), OSI-027, AZD2014, CC-223, RADOOl, LY294002, BEZ235, rapamycin, Ku-0063794, or PP242. Inhibitors of AKT include MK-2206, GDC-0068 and GSK795. Inhibitors of MEK includes trametinib, selumetinib, cobimetinib, MEK162, PD-325901, PD-035901, AZD6244, and CI-1040. The application also uses and includes other inhibitors, such as CDK inhibitors (AT-7519, SNS- 032), JNK inhibitors (CC-401), MAPK inhibitors (VX-702, SB203580, SB202190), Raf inhibitors (PLX4720), Tie2 inhibitor (AMG-Tie2-1). As described herein, such inhibitors include compounds or agents that inhibit all subclasses (e.g., isoforms or members) of a target (e.g. PI3K alpha, beta, delta and gamma), compounds or agents that inhibit primarily one subclass, and compounds or agents that inhibit a subset of all subclasses.
In the present application, the one or more therapeutic agents, including the ROCK inhibitor and/or JAK inhibitor, may be used or combined with a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof. In some embodiments, the one or more therapeutic agents are compounds or molecules that is an Abl inhibitor, an ACK inhibitor, an A2B inhibitor, an ASK inhibitor, an Aurora kinase inhibitor, a BTK inhibitor, a BRD inhibitor, a BCL2 inhibitor, a c- Kit inhibitor, a c-Met inhibitor, a CAK inhibitor, a CaMK inhibitor, a CDK inhibitor, a CK inhibitor, a DDR inhibitor, an EGFR inhibitor, a FAK inhibitor, a Flt-3 inhibitor, a FYN inhibitor, a GSK inhibitor, a HCK inhibitor, a HDAC inhibitor, an IKK inhibitor, an IDH inhibitor, an IKK inhibitor, a JAK inhibitor, a KDR inhibitor, a LCK inhibitor, a LOX inhibitor, a LOXL inhibitor, a LYN inhibitor, a MMP inhibitor, a MEK inhibitor, a MARK inhibitor, a MDM2 inhibitor, a NEK9 inhibitor, a NPM- ALK inhibitor, a p38 kinase inhibitor, a PDGF inhibitor, a PI3 kinase (PI3K), a PK inhibitor, a PLK inhibitor, a PK inhibitor, a PYK inhibitor, a SYK inhibitor, a TPL2 inhibitor, a STK inhibitor, a STAT inhibitor, a SRC inhibitor, a TB K inhibitor, a TIE inhibitor, a TK inhibitor, a telomerase inhibitor, a VEGF inhibitor, a YES inhibitor, a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti-neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti-angiogenic agent, a therapeutic antibody, or any combination thereof.
Chemotherapeutic agents may be categorized by their mechanism of action into, for example, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (floxuridine, capecitabine, and cytarabine); purine analogs, folate antagonists and related inhibitors antiproliferative/antimitotic agents including natural products such as vinca alkaloid (vinblastine, vincristine) and microtubule such as taxane (paclitaxel, docetaxel), vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide); DNA damaging agents (actinomycin, amsacrine, busulfan, carboplatin, chlorambucil, cisplatin, cyclophosphamide, Cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin, iphosphamide, melphalan, merchlorehtamine, mitomycin, mitoxantrone, nitrosourea, procarbazine, taxol, taxotere, teniposide, etoposide, triethylenethiophosphoramide); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L- asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards cyclophosphamide and analogs, melphalan, chlorambucil), and (hexamethylmelamine and thiotepa), alkyl nitrosoureas (BCNU) and analogs, streptozocin), trazenes-dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, oxiloplatinim, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel; antimigratory agents; antisecretory agents (breveldin); immunosuppressives tacrolimus sirolimus azathioprine, mycophenolate; compounds (TNP-470, genistein) and growth factor inhibitors (vascular endothelial growth factor inhibitors, fibroblast growth factor inhibitors); angiotensin receptor blocker, nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab, rituximab); cell cycle inhibitors and differentiation inducers (tretinoin); inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin, irinotecan and mitoxantrone, topotecan, irinotecan), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone); growth factor signal transduction kinase inhibitors; dysfunction inducers, toxins such as Cholera toxin, ricin, Pseudomonas exotoxin, Bordetella pertussis adenylate cyclase toxin, or diphtheria toxin, and caspase activators; and chromatin.
As used herein the term "chemotherapeutic agent" or "chemotherapeutic" (or "chemotherapy," in the case of treatment with a chemotherapeutic agent) is meant to encompass any non-proteinaceous (i.e, non-peptidic) chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN(tm)); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; emylerumines and memylamelamines including alfretamine, triemylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimemylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (articularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, foremustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin phill, see, e.g., Agnew, Chem. Inti. Ed. Engl, 33:183-186 (1994); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzi nostatin chromophore and related chromoprotein enediyne antibiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carrninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (Adramycin™) (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as demopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogues such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replinisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; hestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformthine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; leucovorin; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; losoxantrone; fluoropyrimidine; folinic acid; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK(r); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-tricUorotriemylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethane; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiopeta; taxoids, e.g., paclitaxel (TAXOL®), Bristol Meyers Squibb Oncology, Princeton, N.J.) and docetaxel (TAXOTERE(r), Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine (Gemzar®); 6- thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitroxantrone; vancristine; vinorelbine (Navelbine(r)); novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeoloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; FOLFIRI (fluorouracil, leucovorin, and irinotecan) and pharmaceutically acceptable salts, acids or derivatives of any of the above.
Also included in the definition of "chemotherapeutic agent" are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including Nolvadex™), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston®); inhibitors of the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate (Megace®), exemestane, formestane, fadrozole, vorozole (Rivisor®), letrozole (Femara®), and anastrozole (Arimidex®); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprohde, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. The anti-angiogenic agents include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATIN®, ENDOSTATIN®, suramin, squalamine, tissue inhibitor of metalloproteinase-1, tissue inhibitor of metalloproternase-2, plasminogen activator inhibitor-1, plasminogen activator inbibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulphate (clupeine), sulphated chitin derivatives (prepared from queen crab shells), sulphated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-azetidine-2-carboxylic acid (LACA), cishydroxyproline, d,l-3,4-dehydroproline, thiaproline, .alpha. -dipyridyl, beta-aminopropionitrile fumarate, 4-propyl-5-(4-pyridinyl)-2(3h)-oxazolone; methotrexate, mitoxantrone, heparin, interferons, 2 macroglobulin-serum, chimp-3, chymostatin, beta-cyclodextrin tetradecasulfate, eponemycin; fumagillin, gold sodium thiomalate, d-penicillamine (CDPT), beta-1- anticollagenase-serum, alpba-2-antiplasmin, bisantrene, lobenzarit disodium, n-2- carboxyphenyl-4-chloroanthronilic acid disodium or "CCA", thalidomide; angiostatic steroid, cargboxynaminolmidazole; metalloproteinase inhibitors such as BB94. Other anti-angiogenesis agents include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: beta-FGF, alpha-FGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-l/Ang-2. See Ferrara N. and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.
The anti-fibrotic agents include, but are not limited to, the compounds such as beta- aminoproprionitrile (BAPN), as well as the compounds disclosed in U.S. Pat. No. 4,965,288 to Palfreyman, et al., issued Oct. 23, 1990, entitled "Inhibitors of lysyl oxidase," relating to inhibitors of lysyl oxidase and their use in the treatment of diseases and conditions associated with the abnormal deposition of collagen; U.S. Pat. No. 4,997,854 to Kagan, et al., issued Mar. 5, 1991, entitled "Anti-fibrotic agents and methods for inhibiting the activity of lysyl oxidase in situ using adjacently positioned diamine analogue substrate," relating to compounds which inhibit LOX for the treatment of various pathological fibrotic states, which are herein incorporated by reference. Further exemplary inhibitors are described in U.S. Pat. No. 4,943,593 to Palfreyman, et al., issued Jul. 24, 1990, entitled "Inhibitors of lysyl oxidase," relating to compounds such as 2-isobutyl-3- fluoro-, chloro-, or bromo-allylamine; as well as, e.g., U.S. Pat. No. 5,021,456; U.S. Pat. No. 5,5059,714; U.S. Pat. No. 5,120,764; U.S. Pat. No. 5,182,297; U.S. Pat. No. 5,252,608 (relating to 2-(l-naphthyloxymemyl)-3-fluoroallylamine); and U.S. Patent Application No. 2004/0248871, which are herein incorporated by reference. Exemplary anti-fibrotic agents also include the primary amines reacting with the carbonyl group of the active site of the lysyl oxidases, and more particularly those which produce, after binding with the carbonyl, a product stabilized by resonance, such as the following primary amines: emylenemamine, hydrazine, phenylhydrazine, and their derivatives, semicarbazide, and urea derivatives, aminonitriles, such as betaaminopropionitrile (BAPN), or 2-nitroethylamine, unsaturated or saturated haloamines, such as 2-bromo-ethylamine, 2-chloroethylamine, 2-trifluoroethylamine, 3-bromopropylamine, p- halobenzylamines, selenohomocysteine lactone. Also, the anti-fibrotic agents are copper chelating agents, penetrating or not penetrating the cells. Exemplary compounds include indirect inhibitors such compounds blocking the aldehyde derivatives originating from the oxidative deamination of the lysyl and hydroxylysyl residues by the lysyl oxidases, such as the thiolamines, in particular D-penicillamine, or its analogues such as 2-amino-5-mercapto-5-methylhexanoic acid, D-2-amino-3-methyl-3-((2-acetamidoethyl)dithio)butanoic acid, p-2-amino-3-methyl-3-((2- aminoethyl)dithio)butanoic acid, sodium-4-((p-l-dimethyl-2-amino-2-carboxyethyl)dithio) butane sulphurate, 2-acetamidoethyl-2-acetamidoethanethiol sulphanate, sodium-4-mercapto- butanesulphinate trihydrate.
The immunotherapeutic agents include and are not limited to therapeutic antibodies suitable for treating patients; such as abagovomab, adecatumumab, afutuzumab, alemtuzumab, altumomab, amatuximab, anatumomab, arcitumomab, bavituximab, bectumomab, bevacizumab, bivatuzumab, blinatumomab, brentuximab, cantuzumab, catumaxomab, cetuximab, citatuzumab, cixutumumab, clivatuzumab, conatumumab, daratumumab, drozitumab, duligotumab, dusigitumab, detumomab, dacetuzumab, dalotuzumab, ecromeximab, elotuzumab, ensituximab, ertumaxomab, etaracizumab, farietuzumab, ficlatuzumab, figitumumab, flanvotumab, futuximab, ganitumab, gemtuzumab, girentuximab, glembatumumab, ibritumomab, igovomab, imgatuzumab, indatuximab, inotuzumab, intetumumab, ipilimumab, iratumumab, labetuzumab, lexatumumab, lintuzumab, lorvotuzumab, lucatumumab, mapatumumab, matuzumab, milatuzumab, minretumomab, mitumomab, moxetumomab, narnatumab, naptumomab, necitumumab, nimotuzumab, nofetumomabn, ocaratuzumab, ofatumumab, olaratumab, onartuzumab, oportuzumab, oregovomab, panitumumab, parsatuzumab, patritumab, pemtumomab, pertuzumab, pintumomab, pritumumab, racotumomab, radretumab, rilotumumab, rituximab, robatumumab, satumomab, sibrotuzumab, siltuximab, simtuzumab, solitomab, tacatuzumab, taplitumomab, tenatumomab, teprotumumab, tigatuzumab, tositumomab, trastuzumab, tucotuzumab, ublituximab, veltuzumab, vorsetuzumab, votumumab, zalutumumab, CC49 and 3F8. The exemplified therapeutic antibodies may be further labeled or combined with a radioisotope particle, such as indium In111, yttrium Y90, iodine I131.
In one embodiment, the one or more additional therapeutic agent may be an inhibitor to PI3K such as PI3Ky, PI3K6, PI3K0, and PI3Ka, JAK such as JAK1 and JAK2, SYK, BTK, BRD such as BRD4, BCL2 inhibitor, lysyl oxidase protein, lysyl oxidase-like protein (LOXL) such as LOXL1, LOXL2, LOXL3, LOXL4, or LOXLS, matrix metalloprotease (MMP) such as MMP 1-10, adenosine A2B receptor (A2b), isocitrate dehydrogenase (IDH) such as IDH1, apoptosis signal-regulating kinase (ASK) such as ASK1, serine/threonine kinase TPL2, discoidin domain receptor (DDR) such as DDR1 and DDR2, histone deacetylase (HDAC) inhibitor protein kinase C (PKC) or any combination thereof. In otherembodiment, the one or more therapeutic agent may be a PI3K (including PI3 Ky, PI3K6, PI3K0, PI3Ka, and/or pan-PI3K) inhibitor, a JAK (including JAK1 and/or JAK2) inhibitor, a SYK inhibitor, a BTK inhibitor, an A2B (adenosine A2B receptor) inhibitor, an ACK (activated CDC kinase, including ACK1) inhibitor, an ASK (apoptosis signal-regulating kinase, including ASK1) inhibitor, Aurora kinase, a BRD (bromodomain-containing protein, including BRD4) inhibitor, a CAK (CDK-activating kinase) inhibitor, a CaMK (calmodulin-dependent protein kinases) inhibitor, a CDK (cyclin-dependent kinases, including CDK1, 2, 3, 4, and/or 6) inhibitor, a CK (casein kinase, including CK1 and/or CK2) inhibitor, a DDR (discoidin domain receptor, including DDR1 and/or DDR2) inhibitor, a EGFR inhibitor, a FAK (focal adhesion kinase) inhibitor, a GSK (glycogen synthase kinase) inhibitor, a HDAC (histone deacetylase) inhibitor, an IDH (isocitrate dehydrogenase, including IDH1) inhibitor, an IKK inhibitor, a LCK (lymphocyte-specific protein tyrosine kinase) inhibitor, a LOX (lysyl oxidase) inhibitor, a LOXL (lysyl oxidase like protein, including L0XL1, L0XL2, L0XL3, L0XL4, and/or L0XL5) inhibitor, a MEK inhibitor, a matrix metalloprotease (MMP, including MMP2 and/or MMP9) inhibitor, a mitogen-activated protein kinases (MAPK) inhibitor, a MDM2 inhibitor, a PDGF (platelet-derived growth factor) inhibitor, a phosphorylase kinase (PK) inhibitor, a PLK (polo-like kinase, including PLK1, 2, 3) inhibitor, a protein kinase (PK, including protein kinase A, B, C) inhibitor, a serine/threonine kinase (STK) inhibitor, a STAT (signal transduction and transcription) inhibitor, a TBK (serine/threonine- protein kinase, includin TBK1) inhibitor, a TK (tyrosine kinase) inhibitor, a TPL2 (serine/threonine kinase) inhibitor, a telomerase inhibitor, a NEK9 inhibitor, an Abl inhibitor, a p38 kinase inhibitor, a PYK inhibitor, a PYK inhibitor, a c-Kit inhibitor, a NPM-ALK inhibitor, a Flt-3 inhibitor, a c-Met inhibitor, a KDR inhibitor, a TIE-2 inhibitor, a VEGFR inhibitor, a SRC inhibitor, a HCK inhibitor, a LYN inhibitor, a FYN inhibitor, a YES inhibitor, or any combination thereof.
In other embodiments, the one or more therapeutic agent is: a JAK inhibitor, including but not limited to ruxolitinib, fedratinib, tofacitinib, baricitinib, lestaurtinib, pacritinib, XL019, AZD1480, INCB039110, LY2784544, BMS911543, and NS018; a myelofibrosis inhibiting agent, including but not limited to, hedgehog inhibitors (saridegib), histone deacetylase (HDAC) inhibitors (pracinostat, panobinostat), tyrosine kinase inhibitor (lestaurtinib); a discoidin domain receptor (DDR) inhibitor, including but not limited to, those disclosed in US2009/0142345, US2011/0287011, W02013/027802, WO2013/034933, and U.S. Provisional Application No. 61/705,044; a MMP9 inhibitor, including but not limited to, marimastat (BB-2516), cipemastat (Ro 32-3555), and those described in WO2012/027721; a LOXL inhibitor, including but not limited to the antibodies described in W02009/017833; a L0XL2 inhibitor, including but not limited to the antibodies described in W02009/017833, W02009/035791 and WO/2011/097513; an ASK1 inhibitor, including but not limited to, those described in W02011/008709 and WO/2013/112741; a PI3K6 inhibitor, described in U.S. Pat. No. 7,932,260, U.S. Provisional Application Nos. 61/745,437 and 61/835,333, PI3K II, TGR-1202, AMG-319, GSK2269557, X-339, X-414, RP5090, KAR4141, XL499, OXY111A, IPI-145, IPI-443; a PI3K inhibitor, including but not limited to, GSK2636771, BAY 10824391, TGX221; a PI3Ka inhibitor, including but not limited to, Buparlisib, BAY 80-6946, BYL719, PX-866, RG7604, MLN1117, WX-037, AEZS-129, PA799; a PI3Ky inhibitor, including but not limited to, ZSTK474, AS252424, LY294002, TG100115; a pan PI3K inhibitor, including but not limited to, LY294002, BEZ235, XL147 (SAR245408), GDC-0941; additional PI3K inhibitors, including but not limited to BKM120, CH5132799, XL756, and GDC- 0980, wortmannin; a BTK inhibitor, including but not limited to, ibrutinib, HM71224, ONO-4059, CC-292; a BCL2 inhibitor including but not limited to venetoclax, navitoclax, a SYK inhibitor, including but not limited to, tamatinib (R406), fostamatinib (R788), PRT062607, BAY-61-3606, NVP-QAB 205 AA, R112, R343, or those described in U.S. Pat. No. 8,450,321; a BRD4 inhibitor, an IDH1 inhibitor, a TPL2 inhibitor, an A2b inhibitor, or agents that activate or reactivate latent human immunodeficiency virus (HIV), or a protein kinase C (PKC) activator, romidepsin or panobinostat. In another embodiment, JAK inhibitors include, but not limited to, Decernotinib (or VX-509), GLPG0634, or GLPG0788, a MDM2 inhibitors include, but not limited to RG7112 (RO5045337), idasanutlin (RG7388), AMG-232 (KRT-232), APG-115, BI-907828, CGM097, siremadlin (HDM201), and milademetan (DS-3032b), telomerase inhibitors include, but not limited to imetelstat, or a pharmaceutically acceptable salt thereof.
In other embodiments, the one or more therapeutic agent is selected from the following: interferon alpha ligand modulators, such as interferon alpha-2b, interferon alpha-2a biosimilar (Biogenomics), ropeginterferon alfa-2b (AOP-2014, P-1101, PEG IFN alpha-2b), Multiferon (Alfanative, Viragen), interferon alpha lb, Roferon-A (Canferon, Ro-25-3036), interferon alfa-2a follow-on biologic (Biosidus) (Inmutag, Inter 2A), interferon alfa-2b follow-on biologic (Biosidus— Bioferon, Citopheron, Ganapar, Beijing Kawin Technology— Kaferon), Alfaferone, pegylated interferon alpha-lb, peginterferon alfa-2b follow-on biologic (Amega), recombinant human interferon alpha-lb, recombinant human interferon alpha-2a, recombinant human interferon alpha-2b, veltuzumab-IFN alpha 2b conjugate, Dynavax (SD-101), and interferon alfa-nl (Humoferon, SM-10500, Sumiferon); interferon gamma ligand modulators, such as interferon gamma (OH-6000, Ogamma 100).
The Compounds for Treating Myeloproliferative Disorders and Neoplasms.
In a first aspect of the present invention, there is provided a compound of Formula (I) for treating a myeloproliferative disease or disorder, or for treating a disease or condition associated with myeloproliferative neoplasms:
Figure imgf000027_0001
' , wherein
Zi is OR', phenyl, naphthyl, or C5-C10 membered heterocycle, wherein the phenyl, naphthyl, or C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10- membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 10- membered heterocycle is optionally substituted with one or more of the following: halo, -OH, - CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR’R”, -C(O)NR’R”, - NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl;
Z2 is phenyl, naphthyl, or C5-C10 membered heterocycle, wherein the phenyl, naphthyl, or C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -OR'OR", -O(CH2)2NR'R", -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", - NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 10- membered heterocycle is optionally substituted with one or more of the following: halo, -OH, - CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl;
Ri is H, -C1-C6 alkyl or -C3-C7 cycloalkyl, wherein the -C1-C6 alkyl or -C3-C7 cycloalkyl is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", or - S(O)2R';
R is -C1-C6 alkyl, wherein the -C1-C6 alkyl is optionally substituted with one or more of the following: H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", or -S(O)2R';
X is a bond or -OCH(R4), wherein R4 is H, -C1-C6 alkyl or -C3-C7 cycloalkyl; R2 and R3 are independently H, -C1-C6 alkyl, -C3-C7 cycloalkyl, aryl, C5-C10 membered
Figure imgf000028_0001
heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle, wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -CH2NR'R", -OR', -OR'R", - SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", - S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, and wherein R5 is -CI-C6 alkyl, -OCH2CH2-, -NR6CH2CH2-, -NC(O)CH2CH2-; R6 is H, -C1-C6 alkyl or -C3-C7 cycloalkyl; and
R' or R" is independently -H or -CI-C6 alkyl, or R' and R" together, optionally attaching to N or O atom, form a 4- to 8-membered cyclic structure.
In some non-limiting embodiments, Zi is an optionally substituted C5-C10 membered heterocycle. In certain non-limiting implementations, Zi is an optionally substituted pyridine, pyrimidine, pyrazole, imidazole, oxazole, thiazole, indazole or tetrazole. In further non-limiting implementations, Zi is an optionally substituted pyridine or pyrazole. The C5-C10 membered heterocycle, pyridine, pyrimidine, pyrazole, imidazole, oxazole, thiazole, indazole or tetrazole is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 10-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl. In other non-limiting embodiments, Zi is an unsubstituted C5-C10 membered heterocycle. In certain non-limiting implementations, Zi is an unsubstituted pyridine, pyrimidine, pyrazole, imidazole, oxazole, thiazole, indazole or tetrazole. In further non-limiting implementations, Zi is an unsubstituted pyridine or pyrazole. In yet further non-limiting implementations, Zi is an unsubstituted
Figure imgf000029_0001
. in some embodiments, Zi is -
OR', wherein R' is methyl (i.e., Zi is methoxy).
In some non-limiting embodiments, Z2 is an optionally substituted C5-C10 membered heterocycle. In certain non-limiting implementations, Z2 is an optionally substituted phenyl, pyridine, or pyrazole. The C5-C10 membered heterocycle, phenyl, pyridine, or pyrazole is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered heterocycle. In further non-limiting implementations, Z2 is a phenyl, pyridine or pyrazole optionally substituted with halo, -OR', -Cl- C6 alkyl, -OR'OR", or -O(CH2hNR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R", and wherein the R' or R" are independently -H, methyl or ethyl. In yet further non-limiting implementations, Z2 is phenyl substituted with halo, -OR', -CI-C6 alkyl, - OR'OR", or -O(CH2)2NR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R", and wherein the R' or R" are independently -H, methyl or ethyl.
In other non-limiting embodiments, Z2 is an unsubstituted C5-C10 membered heterocycle. In certain non-limiting implementations, Z2 is an unsubstituted phenyl, pyridine, or pyrazole. In further non-limiting implementations, Z2 is selected from the group consisting of:
Figure imgf000029_0002
In some non-limiting embodiments, Ri is a H, unsubstituted methyl, methoxyethyl or dimethylaminoethyl. In some non-limiting implementations, Ri is H or
Figure imgf000029_0003
In some non-limiting embodiments, R is a methyl. In other non-limiting embodiments, R is hydroxymethyl and the compound has a structure of Formula (III):
Figure imgf000030_0001
111 . In further non-limiting implementations, R is a hydroxymethyl with 5 configuration or a methyl with R configuration. In some non-limiting embodiments, X is a bond.
In other non-limiting embodiments, X is -OCHfRzi), and the compound has a structure of
Figure imgf000030_0002
In some non-limiting embodiments, R2 is H, and R3 is H, -C1-C6 alkyl, -C3-C7 cycloalkyl,
Figure imgf000030_0003
CIO membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle, wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -CH2NR'R", - OR', -OR'R", -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, wherein R5 is - CI-C6 alkyl, -OCH2CH2-, -NR6CH2CH2-, -NC(O)CH2CH2-, and wherein R6 is H, -C1-C6 alkyl or -C3-C7 cycloalkyl.
In certain non-limiting implementations, R2 is H, and R3 is -C3-C7 cycloalkyl or -C3-C7 cycloalkyl methyl, wherein the -C3-C7 cycloalkyl or -C3-C7 cycloalkyl methyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle.
Figure imgf000031_0001
In further non-limiting implementations, R2 is H, and R3 is O
Figure imgf000031_0002
cycloalkyl selected from the group consisting of: cyclohexyl, cyclopentyl, cyclobutyl, cyclopropyl, cyclohexyl methyl, cyclopentyl methyl, cyclobutyl methyl, cyclopropyl methyl; an aryl selected from the group consisting of: phenyl and benzyl; or a C5-C10 membered heterocycle selected from the group consisting of: pyrrolidi ne-3-yl, piperidine-4-yl, (pyrrolidine-3-yl)methyl, (piperidine-4-yl)methyl, wherein the cyclohexyl, cyclopentyl, cyclobutyl, cyclopropyl, cyclohexyl methyl, cyclopentyl methyl, cyclobutyl methyl cyclopropyl methyl phenyl, benzyl, pyrrolidine-3-yl, piperidine-4-yl, (pyrrolidine-3-yl)methyl or (piperidine-4- yl)methyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', - NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle. In yet further non-limiting implementations, R2 is H, and R3 is cyclopentyl or cyclohexyl.
In other non-limiting embodiments, R2 and R3 are independently H, -C3-C7 cycloalkyl, or -C3-C7 cycloalkyl methyl; wherein the -C3-C7 cycloalkyl or -C3-C7 cycloalkyl methyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 11-membered heterocycle; with the proviso that R2 and R3 are not both H. In certain non-limiting implementations, R2 and R3 are independently H, cyclohexyl, cyclopentyl, cyclobutyl, cyclopropyl, cyclohexyl methyl, cyclopentyl methyl, cyclobutyl methyl or cyclopropyl methyl; and R2 and R3 are independently optionally substituted with H, halo, -OH, - CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 11-membered heterocycle.
In other non-limiting implementations, R2 and R3 are independently H, phenyl, or benzyl; wherein the phenyl or benzyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 11-membered heterocycle; with the proviso that R2 and R3 are not both H.
In some embodiments, Ri, R2, or both are H.
In certain non-limiting implementations, Zi is a pyridine or pyrazole; Z2 is an unsubstituted phenyl, pyridine, or pyrazole, or a phenyl substituted with halo, -OR', -CI-C6 alkyl, -OR'OR", or - O(CH2)2NR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R"; Ri is H, unsubstituted methyl, or dimethylamine; X is a bond; R2 is H; and the R' or R" is independently -H, methyl or ethyl.
In other non-limiting implementations, Zi is a pyridine, optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl; Z2 is a C5-C10 membered heterocycle, optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", - C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 11-membered heterocycle; R2 and R3 are independently H, phenyl, benzyl, - C3-C7 cycloalkyl, or -C3-C7 cycloalkyl methyl, wherein the phenyl, benzyl, -C3-C7 cycloalkyl or - C3-C7 cycloalkyl methyl is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3 to 11-membered heterocycle; with the proviso that R2 and R3 are not both H. In further embodiments, R is hydroxymethyl and the compound has a structure of
Formula (IV):
Figure imgf000033_0001
Zi is -pyridine, -pyrimidine, -pyrazole, -imidazole, -oxazole, -thiazole, -indazole, or - tetrazole, wherein the -pyridine, -pyrimidine, -pyrazole, -imidazole, -oxazole, -thiazole, -indazole, or -tetrazole is unsubstituted or substituted with one or more of the following: -halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', -guanidino, -nitro, -nitroso, -Ci-Ce alkyl, -aryl, -C3-C7 cycloalkyl, and a 3- to 10-membered heterocycle, and the -Ci-Ce alkyl, -aryl, -C3-C7 cycloalkyl, or 3- to 10-membered heterocycle is unsubstituted or substituted with one or more of the following: -halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', -guanidino, -nitro, -nitroso, -Ci-C6 alkyl, -aryl, and -C3-C7 cycloalkyl;
Ri is -H, -Ci-Ce alkyl, or -C3-C7 cycloalkyl, wherein the -Ci-Ce alkyl or -C3-C7 cycloalkyl is unsubstituted or substituted with one or more of the following: -halo, -OH, -CN, -COOR', -OR', - SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", and -S(O)2R';
R2 and R3 are independently -H, -Ci-Ce alkyl, -C3-C7 cycloalkyl, -aryl, a heterocycle
Figure imgf000033_0002
alkyl, aryl, -C3-C7 cycloalkyl, and 3- to 11-membered heterocycle, wherein the -Ci-Ce alkyl, aryl, - C3-C7 cycloalkyl, or 3- to 11-membered heterocycle is unsubstituted or substituted with one or more of the following: -halo, -CNR'R", -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", - NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -OR'OR", -S(O)2NR'R", -S(O)2R', -guanidino, - nitro, -nitroso, -CI-C6 alkyl, -aryl, and -C3-C7 cycloalkyl; Rs is -Ci-Ce alkyl, -OCH2CH2-, -NR6CH2CH2- , or -NC(O)CH2CH2-, and Re is -H, -Ci-Ce alkyl, or -C3-C7 cycloalkyl;
R' and R" are independently -H or -Ci-Ce alkyl, or R' and R" together, optionally attached to N or O atom, form a 4- to 8- membered cyclic structure; and
R7 is -H, -halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', -Ci-C6 alkyl, -C3-C7 cycloalkyl, -aryl, or a heterocycle comprising 5 to 10 carbons, wherein the -Ci-Ce alkyl, -C3-C7 cycloalkyl, -aryl, or heterocycle comprising 5 to 10 carbons is unsubstituted or substituted with - -halo, -OH, -CN, - COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', -guanidino, -nitro, -nitroso, -Ci-Ce alkyl, -aryl, -C3-C7 cycloalkyl, or 3- to 10- membered heterocycle.
In some implementations, at least one compound for treating a myeloproliferative disorder or neoplasm is selected from the non-limiting group consisting of:
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Table 1. Non-limiting examples of compounds for treating myeloproliferative disorders and neoplasms.
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0002
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Unless specified, example compounds with a chiral center represent a racemic mixture of the corresponding R and S enantiomers, and all racemates and isolated enantiomers are within the scope of the invention. Additional non-limiting examples are:
Figure imgf000057_0001
Figure imgf000058_0001
In certain embodiments, at least one compound is selected from the group consisting of:
Figure imgf000058_0002
Figure imgf000059_0001
Example 1
ROCK and AURK Inhibition Assay
Methods
The following assay protocol is for measuring the phosphorylation of a peptide substrate (FAM-KKLRRTLSVA-OH wherein FAM is carboxyfluorescein). The peptide is >98% purity by Capillary Electrophoresis. The peptide is phosphorylated by the protein kinase ROCK or AURK. The ROCK or AURK enzyme, substrate, and cofactors (ATP and Mg2+) are combined in a well of a microtiter plate and incubated for 3 hours at 25° C. At the end of the incubation, the reaction is quenched by the addition of an EDTA-containing buffer. The substrate and product are separated and quantified electrophoretically using the microfluidic-based LABCHIP® 3000 Drug Discovery System from Caliper Life Sciences (Hopkinton, Mass.).
Substrate and product peptides present in each sample are separated electrophoretically using the LABCHIP® 3000 capillary electrophoresis instrument. As substrate and product peptides are separated two peaks of fluorescence are observed. Change in the relative fluorescence intensity of the substrate and product peaks is the parameter measured reflecting enzyme activity. Capillary electropherograms (RDA acquisition files) are analyzed using HTS Well Analyzer software (Caliper Life Sciences, Hopkinton, Mass.). The kinase activity in each sample is determined as the product to sum ratio (PSR): P/(S+P), where P is the peak height of the product peptide and S is the peak height of the substrate peptide. For each compound, enzyme activity is measured at various concentrations (12 concentrations of compound spaced by 3x dilution intervals). Negative control samples (0%— inhibition in the absence of inhibitor) and positive control samples ( 100%— i nhibition in the presence of 20 mM EDTA) are assembled in replicates of four and are used to calculate %-inhibition values for each compound at each concentration.
Percent inhibition (Pinh) is determined using the following equation: Pinh=(PSR0%-PSRinh)/(PSR0%-PSR100%)*100 where PSRinh is the product sum ratio in the presence of inhibitor, PSR0% is the average product sum ratio in the absence of inhibitor, and PSR100% is the average product sum ratio in 100%-inhibition control samples. The IC50 values of inhibitors are determined by fitting the inhibition curves (Pin/i versus inhibitor concentration) by 4 parameter sigmoidal dose-response model using XLfit 4 software (IBDS). This assay can be used to test the activity of each of the exemplary compounds identified in Table 2.
Table 2
Figure imgf000061_0001
Example 2
Effects on Phosphorylated STAT3 in IL-2 activated human peripheral blood mononuclear cells (PBMC)
Human PBMC were plated at 5.45 in 6 well plates and treated with DMSO, KD025 (lOmM) or CMPD I D#10 (500,200 and lOOnM) for 1 hr. Subsequently cells were treated with IL2 106 U/ml and cell lysates were collected at one hour post IL-2 stimulation. Lysates were prepared in radioimmunoprecipitation assay buffer (RIPA buffer) containing protease and phosphatase inhibitors.
• 10 mg protein lysates were loaded onto a gel, transferred, and blocked.
• Blots were analyzed by specific antibodies to STAT3 and pSTATB.
• Blots were stripped and incubated with specific antibodies to b-actin as a protein loading control.
• Blots were developed by horseradish peroxidase and bands were quantitated using Image Lab software. The bands were normalized to b-actin levels of the DMSO treatment at one hour. Normalized STAT and pSTAT band volumes were used to calculate ratio of phosphorylated to total STAT levels. These ratios were expressed as percentage of the DMSO treatment and presented as bar graphs. See Figures 1, 2. CMPD ID#97 caused a dose dependent inhibition of STAT3 phosphorylation. Inhibition of pSTAT3 phosphorylation with belumosudil (KD025) was observed at lOmM and was comparable to lOOnM of CMPD ID#97. See Figure 2.
Example 3
Effects on Myosin light chain (MLC2) and phosphorylated (pMLC2) in human HEL 92.1.7 and UKE-1 cell lines.
Human Hel 92.1.7 and UKE1 cells were plated at 5.0xl05 cells per well of a 6 well dish. 24 hours after plating, cells were treated with DMSO or CMPD #97 (1.0 mm or 0.1 mM) in duplicate. Cells were collected at 24 and 48 hours and lysates prepared in IX lithium dodecyl sulfate (LDS) buffer (Life Technologies, Carlsbad, CA) supplemented with protease- and phosphatase inhibitors (Roche). Equal volumes of protein were run on 4-12 % gels and transferred to PVDF membranes.
• Blots were blocked in 5% nonfat milk powder and incubated with antibodies against MLC2 or pMLC2 (Cell Signaling Technology) overnight at 4°C.
• Blots were developed with horseradish peroxidase conjugated secondary antibodies and imaged on a MP3 imager (Bio-Rad).
• Blots were stripped and re-probed with b-actin antibodies (Sigma)
• Bands were quantified by Imagelab (Bio-Rad) and normalized to b actin levels of the control at each time point.
• Ratio of phosphorylated MLC2 to MLC2 was calculated and expressed as percentage of the control set to 100%
See Figures 3, 4, 5, 6.
As reflected in Figures 3-6, CMPD #97 significantly decreased phosphorylation of MLC2 (pMLC2) at 1.0 mM and 0.1 mM concentrations, in both human cell lines confirming an on-target inhibition of ROCK. Example 4
Colony Formation Unit (CFU) assay with human HEL92.1.7 and UKE-1 cell lines.
Method:
After premixing 1/100 volume of CMPD #97 DMSO solution with MethoCult H4230 media (StemCell, #04230), 16,500 cells in 0.3 mL cell culture media were added into 3.0 mL MethoCult media in a 5 mL tube. Cells were resuspended into the MethoCult media by gentle agitation. Tubes were kept standing at room temperature for 15 minutes. Cells were placed by a syringe with a blunt end needle into 6-well plates at 5,000 cells/1.0 mL/well. Plates were kept in a cell culture incubator for 7 days. When colony size in control group (DMSO only) reached 50-100 pm, 5-6 images were randomly taken under 4x magnification for each condition, and colony number count by a blinded observer.
The ICso for CMPD I D#97 in human HEL-92.1.7 cells was 25.21 nM. CMPD I D#97 was serially diluted 3:1 from 10 pM (#12) to 0.17 nM (#2). Condition 1 (#1) was DMSO control. See Figure 7. The IC50 for CMPD ID#97 in human UKE-1 cells was 54.62 nM. CMPD I D#97 was serially diluted 3:1 from 10 pM (#9) to 4.57 nM (#2). Condition 1 (#1) was DMSO control. See Figure 8.
Example 5
Cell cycle analysis of human HEL-92.1.7 and UKE-1 cells
Method
The cell lines HEL 92.1.7 and UKE-1 were synchronized with 2mM Thymidine (Cayman Chemical (Ann Arbor, Ml) for 18hr, washed, and rested for4hr. The cells were treated with CMPD #10 and CMPD #97 for 24hr at 500nM and lpM. After treatment, the cells were harvested, washed twice in IX DPBS, and fixed in 70% ice-cold ethanol for lhr on ice. After fixation cells were washed with IX DPBS and let rehydrate for 15 min. A solution of 16.67pg/ml Propidium Iodide (BioLegend (San Diego, CA)), lOOpg/ml RNase A (ThermoFisher (Waltham, MA)), and 0.1% v/v Triton-X 100 (Sigma Aldrich (St Louis, MO)) was used to stain the cells for 15 min at 4°C in the dark.
The samples were analyzed using the MACSQuant Analyzer 16 (Miltenyi Biotec (Bergisch Gladback, Germany)) and a total of 20,000 single cell events were recorded per sample. FlowJo vlO (BD Biosciences (Franklin Lakes, NJ) was used to analyze the data. Doublets were excluded from the analysis by plotting the side scatter width vs. side scatter area. The cell cycle distribution was calculated using the Watson Pragmatic Model. Each condition had two biological replicates that were split into three technical replicates for staining and analysis. See Figures 9, 10, 11.
Example 6
Effects on Myelofibrosis patients Hematopoietic stem and progenitor cells
Methods
1. CMPD ID#97 was added to MethoCult Media (standardized semi-solid methylcellulose media with appropriate cytokines for colony assays) at varying concentrations (0 mM (control), 0.06 mM, 0.19 mM, and 0.56 mM).
2. Peripheral blood mononuclear cells from three healthy donors (HD1, HD2, and HD3) and two myelofibrosis patients (MF2, and MF3) were mixed with the CMPD ID#97- containing MethoCult Media (3,000 cells per mL) and plated into 35-mm dishes.
3. The CFU assay was allowed to grow for 12 days in a humidified incubator at 37°C, 5% CO2.
4. At the end of the culture period, colonies were identified and counted for each CFU type (burst forming colonies of erythrocytes (BFU), granulocytes and macrophages (GM), and granulocytes, erythrocytes, macrophages, and megakaryocytes (GEMM).
See Figures 12, 13.
Example 7
In vivo HEL-92.1.7 Human Erythroleukemia Intravenous Dissemination xenograft
Methods
Six-week-old female CIEA NOG mice (NOD.Cg-Prkdcscid H2rgtmlSug/JicTac), received from Taconic, were inoculated with human HEL-92.1.7 (7.5xl05 cells/mouse) intravenously using a saline vehicle. Mice were randomized into vehicle and treatment groups by body weights five days following tumor cell inoculation and treatment was initiated. Experimental test article CMPD #97 (in 10% DMA: 90% PEG400 vehicle) was administered orally daily at 5 and 25 mg/kg. Ruxolitinib (in 10% DMA: 90% PEG400 vehicle) was also administered orally daily at 45 mg/kg alone or in combination with CMPD #97. Body weights were measured thrice weekly. Study endpoint was time to peri-morbidity to assess tumor growth delay of treated groups compared to vehicle control. See Figure 14.
Unless defined otherwise, all technical and scientific terms herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications, patents, and patent publications cited are incorporated by reference herein in their entirety for all purposes.
It is understood that the disclosed invention is not limited to the particular methodology, protocols and materials described as these can vary. It is also understood that the terminology used herein is for the purposes of describing particular embodiments only and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

CLAIMS What is claimed is:
1. A compound of Formula (I) for use in treating a myeloproliferative disorder or neoplasm in a subject:
Figure imgf000066_0001
, wherein:
Zi is OR', phenyl, naphthyl, or C5-C10 membered heterocycle, wherein the phenyl, naphthyl, or C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', - OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or a 3- to 10-membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 10-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", - S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl;
Z2 is phenyl, naphthyl, or C5-C10 membered heterocycle, wherein the phenyl, naphthyl, or C5- C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', - OR'OR", -O(CH2)2NR'R", -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", - C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3- C7 cycloalkyl, or a 3- to 10-membered heterocycle, and wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 10-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', - OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", - S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, or -C3-C7 cycloalkyl;
Ri is H, -C1-C6 alkyl or -C3-C7 cycloalkyl, wherein the -C1-C6 alkyl or -C3-C7 cycloalkyl is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", or -
S(O)2R';
R is -C1-C6 alkyl, wherein the -C1-C6 alkyl is optionally substituted with one or more of the following: H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', - NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", or -S(O)2R';
X is a bond or -OCH(R4), wherein R4 is H, -C1-C6 alkyl or -C3-C7 cycloalkyl;
R2 and R3 are independently H, -C1-C6 alkyl, -C3-C7 cycloalkyl, aryl, C5-C10 membered
Figure imgf000067_0001
the -C1-C6 alkyl, -C3-C7 cycloalkyl, aryl, C5-C10 membered heterocycle is optionally substituted with H, halo, -OH, -CN, -COOR', -OR', -SR', -OC(O)R', -NHR', -NR'R", -CNR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', -S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle, wherein the -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, or 3- to 11-membered heterocycle is optionally substituted with one or more of the following: halo, -OH, -CN, -COOR', -CH2NR'R", -OR', - OR'R", -SR', -OC(O)R', -NHR', -NR'R", -NHC(O)R', -NHC(O)NR'R", -C(O)NR'R", -NS(O)2R', - S(O)2NR'R", -S(O)2R', guanidino, nitro, nitroso, -CI-C6 alkyl, aryl, -C3-C7 cycloalkyl, and wherein
R5 is -CI-C6 alkyl, -OCH2CH2-, -NR6CH2CH2-, -NC(O)CH2CH2-; R6 is H, -C1-C6 alkyl or -C3-C7 cycloalkyl; and
R' or R" is independently -H or -CI-C6 alkyl, or R' and R" together, optionally attaching to N or O atom, form a 4- to 8-membered cyclic structure.
2. The compound of Claim 1, wherein Zi is pyridine or pyrazole.
The compound of Claim 2, wherein Zi is
Figure imgf000068_0001
4. The compound of Claim 1, wherein Z2 is phenyl, pyridine, or pyrazole, optionally substituted with halo, -OR', -CI-C6 alkyl, -OR'OR", -O(CH2)2NR'R", wherein the -CI-C6 alkyl is optionally substituted with one or more of -OR' or NR'R", and wherein the R' or R" are independently -H, methyl or ethyl.
5. The compound of Claim 4, wherein Z2 is selected from the group consisting of:
Figure imgf000068_0002
6. The compound of Claim 1, wherein Ri is H, unsubstituted methyl, methoxyethyl or dimethylaminoethyl.
7. The compound of Claim 6, wherein Ri is H.
8. The compound of Claim 1, wherein R is methyl or hydroxymethyl.
9. The compound of Claim 8, wherein R is hydroxymethyl with S configuration or methyl with R configuration.
10. The compound of Claim 1, wherein X is a bond.
11. The compound of claim 1, wherein the compound is selected from the group consisting of
Figure imgf000068_0003
Figure imgf000069_0001
12. The compound of claim 3, wherein the compound is selected from the group consisting of
Figure imgf000070_0001
13. The compound of claim 12, wherein the compound is
Figure imgf000070_0002
14. A method of treating a disease or condition associated with myeloproliferative neoplasms, the method comprising administration of an effective amount of a treating agent including a compound of any of claims 1-13.
15. The method of claim 14, wherein the method comprises co-administration of at least one additional treating agent.
16. The method of claim 15, wherein the at least one additional treating agent is selected from the group consisting of compounds or molecules that target a PI3 kinase (PI3K), a spleen tyrosine kinase (SYK), a Janus kinase (JAK), a bromodomain-containing (BRD), a Bruton's tyrosine kinase (BTK), a BCL2 inhibitor, a MDM2 inhibitor, a telomerase inhibitor, an interferon alpha ligand modulator, and a ROCK inhibitor.
17. The method of claim 14, wherein the myeloproliferative disorder is selected from the group consisting of myelofibrosis (MF), polycythemia vera (PV), primary myelofibrosis (PMF), thrombocythemia, essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), postpolycythemia vera myelofibrosis, post-essential thrombocythemia myelofibrosis, chronic myelogenous leukemia (CML), systemic mastocystosis (SM), chronic neutrophilic leukemia (CNL), myelodysplastic syndrome (MDS), systemic mast cell disease (SMCD).
18. The method of claim 17, wherein the method comprises co-administration of at least one or more additional therapeutic agents selected from the group consisting of: a chemotherapeutic agent, an immunotherapeutic agent, a radiotherapeutic agent, an anti- neoplastic agent, an anti-cancer agent, an anti-proliferation agent, an anti-fibrotic agent, an anti- angiogenic agent, and a therapeutic antibody.
19. A kit for treating a disease or condition associated with myeloproliferative neoplasms, the kit comprising the compound of any of claims 1-13.
PCT/US2023/030634 2022-08-19 2023-08-18 Compositions and methods to treat myeloproliferative disorders and myeloproliferative neoplasms Ceased WO2024039883A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263399616P 2022-08-19 2022-08-19
US63/399,616 2022-08-19

Publications (2)

Publication Number Publication Date
WO2024039883A2 true WO2024039883A2 (en) 2024-02-22
WO2024039883A3 WO2024039883A3 (en) 2024-03-28

Family

ID=89942213

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/030634 Ceased WO2024039883A2 (en) 2022-08-19 2023-08-18 Compositions and methods to treat myeloproliferative disorders and myeloproliferative neoplasms

Country Status (1)

Country Link
WO (1) WO2024039883A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12414950B1 (en) 2024-05-21 2025-09-16 Telios Pharma Inc. Methods of treating indolent systemic mastocytosis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10738007B2 (en) * 2016-10-24 2020-08-11 Translation Drug Development, LLC Amide compounds as kinase inhibitors, compositions and methods of treatment
WO2020142745A1 (en) * 2019-01-04 2020-07-09 Translational Drug Development, Llc Methods of treating graft versus host disease and neoplastic disease with amide compounds
CA3184633A1 (en) * 2020-06-03 2021-12-09 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12414950B1 (en) 2024-05-21 2025-09-16 Telios Pharma Inc. Methods of treating indolent systemic mastocytosis

Also Published As

Publication number Publication date
WO2024039883A3 (en) 2024-03-28

Similar Documents

Publication Publication Date Title
US20160279135A1 (en) Therapies for treating myeloproliferative disorders
EP3355875B1 (en) Combination of a btk inhibitor and a checkpoint inhibitor for treating cancers
Druker Imatinib alone and in combination for chronic myeloid leukemia
AU2015206194A1 (en) Therapies for treating cancers
AU2017305303B2 (en) Cobicistat for use in cancer treatments
CN110325191A (en) With the cancer of less side effect treatment EGFR- driving
US20180117052A1 (en) Combination therapies for treating cancers
UA125216C2 (en) COMBINED THERAPY
JP2022538817A (en) Pharmaceutical composition for treating acute myeloid leukemia comprising FLT3 inhibitor and chemotherapeutic agent
US11571400B2 (en) ACAT1 inhibitors for treating cancer
WO2024039883A2 (en) Compositions and methods to treat myeloproliferative disorders and myeloproliferative neoplasms
AU2016304172A1 (en) Combination therapies for treating cancers
NZ750372B2 (en) Cobicistat for use in cancer treatments
OA18380A (en) Combination therapies for treating cancers.

Legal Events

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

Ref document number: 23855523

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 23855523

Country of ref document: EP

Kind code of ref document: A2