WO2020019107A1 - IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT - Google Patents

IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT Download PDF

Info

Publication number
WO2020019107A1
WO2020019107A1 PCT/CN2018/096613 CN2018096613W WO2020019107A1 WO 2020019107 A1 WO2020019107 A1 WO 2020019107A1 CN 2018096613 W CN2018096613 W CN 2018096613W WO 2020019107 A1 WO2020019107 A1 WO 2020019107A1
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitors
group
agents
alkyl
cancer
Prior art date
Application number
PCT/CN2018/096613
Other languages
French (fr)
Inventor
Qingping Zeng
John Patterson
Stephanie Greene
Original Assignee
Fosun Orinove Pharmatech, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fosun Orinove Pharmatech, Inc. filed Critical Fosun Orinove Pharmatech, Inc.
Priority to PCT/CN2018/096613 priority Critical patent/WO2020019107A1/en
Priority to KR1020217004949A priority patent/KR20210036374A/en
Priority to CN201980063917.7A priority patent/CN113164457A/en
Priority to PCT/CN2019/097291 priority patent/WO2020020155A1/en
Priority to AU2019311031A priority patent/AU2019311031A1/en
Priority to US17/261,413 priority patent/US20210260069A1/en
Priority to EP19840929.4A priority patent/EP3826634A4/en
Priority to JP2021503583A priority patent/JP7468829B2/en
Priority to CA3106731A priority patent/CA3106731A1/en
Publication of WO2020019107A1 publication Critical patent/WO2020019107A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • a pharmaceutical combination comprising an IRE1 ⁇ inhibitor and one or more additional cancer therapeutic agents for the treatment of cancerous tumor, a pharmaceutical composition containing the same and a method for treating cancerous tumor using the same.
  • Cancer or cancerous tumor is the second leading cause of death in the developed world and is expected to kill more than 500,000 people in the United States this year.
  • new therapies and improved outcomes new treatments are still required to improve patients’ lives.
  • One such area to this end is using combination therapies to target cancer from multiple weak points or multiple oncogenic drivers.
  • cancer responds to treatments initially but the cancer reoccurs due to resistance and renewal of cancer stem cell survivors.
  • Surgery, chemotherapy and radiotherapy the traditional anti-cancer methods which may not result in complete responses or “cures” , can now be combined with targeted therapies and immunotherapies to improve patient survival outcomes versus using them as single agents.
  • new and novel combinations are desirable to achieve greater therapeutic successes and improved patient outcomes.
  • the tumor microenvironment represents an underutilized therapeutic target area which impacts solid tumor growth and survival.
  • Small molecule modulators of IRE-1 ⁇ kinase and RNase functions have been reported with distinct mechanisms of action reflecting the engagement physically distinct binding sites and direct RNAse active site binding compounds represent a class of modulators that potently, reversibly, and selectively inhibit IRE-1 ⁇ RNase activity including naphthalene (WO 2008/154484 A1; WO 2011/056744 A1) and coumarin (WO 2011/127070 A2) aromatic systems and which may be used as therapeutic agents to treat tumors.
  • naphthalene WO 2008/154484 A1; WO 2011/056744 A1
  • coumarin WO 2011/127070 A2
  • the IRE-1 inhibitor has been studied, as the effective cancer therapy is needed, it is desirable to have effective combined therapy of such inhibitor with other cancer therapeutic agent (s) to maximum the treatment efficacy against various tumors.
  • a pharmaceutical combination comprising
  • R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1 -C 6 hydrocarbon chain containing 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
  • R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino
  • R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6 alkoxyl, C 1-6 hydroxylalkyl, C 1-6 alkoxylC 1-6 alkyl, and
  • R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6 alkoxyl C 1-6 alkyl; or
  • R 9 and R 10 together with the nitrogen atom to which they are attached form 3-10 membered heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6 alkylamino, C 1-6 alkoxyl.
  • the pharmaceutical combination is provided in the form of a pharmaceutical composition.
  • the pharmaceutical combination is provided in the form of one or more kits.
  • a method for treating cancerous tumor comprising administering a subject in need thereof an effective amount of the pharmaceutical combination according to the invention.
  • a method for enhancing the efficacy of a cancer therapeutic agent comprising applying the compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with the cancer therapeutic agent.
  • the compound of formula (I) has the following formula (II) (which is also designated hereinafter as compound Orin1001 or compound 4485) :
  • Figure 1 Induction of percentage of XBP1s relative to total XBP1s and XBP1u measured by RT-qPCR to increasing concentrations of Lestaurtinib (X axis) in MM1s (circles) , HEK293 (triangles) , RPMI 8226 (diamonds) and H929 (squares) cells. Plots were generated using Excel fit software.
  • Figure 2 Nilotinib induced greater than 50 % XBP1s after 2 hours of treatment (triangles) but modest amounts at 1 (circles) or 4 hours (squares) of treatment of MM1s cells.
  • Figure 4 Dasatinib induces highest levels of XBP1s after 1 hour (circles) of treatment.
  • Figure 5 Gefitinib induces highest levels of XBP1s after 2 hours of treatment (triangles) increasing at 1 hour (circles) .
  • Figure 6 Indicated drugs induced potent XBP1s after 1 hour (circles) , 2 hours (triangles) or 4 hours (squares) of treatment shown by IC50 curves for Hepatoma (Hep G2, top panel) MCF-7 (mid panel) and RPMI8226 cells (bottom panel) .
  • Figure 7 Torisel induces high levels of XBP1s after 1 hour (circles) and 4 hours (squares) but modest levels after 2 hours of treatment (triangles) of A549 cells at indicated concentrations.
  • Vorinostat induces high levels of XBP1s after 1 hour (circles) and little after 2 hours (triangles) or 4 hours (squares) of treatment of HT-29 cells at indicated concentrations.
  • Paclitaxel induces high levels of XBP1s after 1 hour (circles) and with modest levels after 4 hours (squares) or 2 hours of treatment (triangles) of RPMI 8226 cells at indicated concentrations.
  • Figure 10 Gemcitabine induces high levels of XBP1s after 4 hours (squares) but low levels after 1 hour (circles) or after 2 hours of treatment (triangles) of RPMI 8226 cells at indicated concentrations.
  • 17-AAG induces high levels of XBP1s after 1 hour (circles) with modest levels after 2 hours (triangles) and low levels after 4 hours (squares) of treatment or of MCF-7 cells at indicated concentrations.
  • 17-AAG induces high levels of XBP1s after 1 hour (circles) with modest levels after 2 hours (triangles) and low levels after 4 hours (squares) of treatment of Hepatoma cells at indicated concentrations.
  • Figure 13 Intratumoral XBP-1 spliced effect of IRE-1 compound Orin1001 in treated RPMI xenografts
  • Figures 14-21 Synergistic effects of compound ORIN 1001 with other cancer therapeutic agents.
  • Figure 22 XBP-1 splicing analysis of liver from compounds dosed PO.
  • proportion is calculated based on weight herein.
  • the term “approximate” or “about” usually refers to the value of the variable and all the values of the variable within the experimental error (for example, within an average 95% confidence interval) or within ⁇ 10% of the specified value, or a wider range.
  • C m-n or “m-n membered” used herein means that the moiety has m-n carbon atoms or m-n atoms.
  • C 1-6 alkyl means said alkyl has 1-6 carbon atoms.
  • C 3-10 cycloalkyl means said cycloalkyl has 3-10 carbon atoms.
  • C m-n is used in a group containing a moiety other than C-containing moiety, it refers to the carbon atom number in said C-containing moiety.
  • C 1-6 in C 1-6 hydroxylalkyl or C 1- 6 alkylamino means that the alkyl therein has 1-6 carbon atoms.
  • C-containing moieties are defined independently, for example C 1-6 alkoxylC 1-6 alkyl. If only one C m-n is defined, it should apply to all C-containing moieties, for example, C 1- 6 alkoxylalkyl means the alkoxyl and alkyl therein are each C 1-6 moiety.
  • C 1-6 means said group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms.
  • C 1-6 alkyl encompasses “C 2-5 alkyl” , “C 1-5 alkyl” , “C 2-6 alkyl” as well as C 1 alkyl, C 2 alkyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, C 6 alkyl or the like.
  • substitution means one or more hydrogen atoms on a given atom are replaced by substituent (s) , provided that the valence of the given atom is normal and the compound after substitution is stable.
  • any variable e.g. R
  • R any variable
  • hetero means heteroatom or heteroatom radical (i.e. a radical containing heteroatom) , i.e. the atoms beyond carbon and hydrogen atoms or the radical containing such atoms.
  • the heteroatom is independently selected from the group consisting of O, N, S and the like.
  • the two or more heteroatoms may be the same, or part or all of the two or more heteroatoms may be different.
  • alkyl refers to a linear or branched saturated aliphatic hydrocarbyl group composed of carbon and hydrogen atoms.
  • Non-limiting examples of C 1-6 alkyl comprise but not limited to methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl or the like.
  • alkoxyl refers to an “alkyl” which is connected to the rest of the molecule via “-O-” , wherein the “alkyl” is defined as above.
  • C 1-6 alkoxyl comprise but not limited to methoxyl, ethoxyl, propoxy or the like.
  • cycloalkyl refers to saturated monocyclic or polycyclic hydrocarbyl group composed of carbon and hydrogen atoms. Cycloalkyl may contain 3-10, for example, 3-8, 3-7, 3-6, 3-5, 4-7, 4-6, or 3-4 carbon atoms or the like, or 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. Non-limiting examples of C 3-10 cycloalkyl comprise but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or the like. The cycloalkyl may further optionally contain one or more (preferably 1 or 2) heteroatoms independently selected from the group consisting of N, O, and S.
  • heterocycle refers to a saturated or unsaturated monocyclic or polycyclic system group, wherein part of the ring atoms (e.g. 1, 2, 3 or 4) are heteroatoms independently selected from the group consisting of N, O and S, and rest of the ring atoms are C.
  • 3-10 membered heterocycle contains 3-10 ring atoms in the system, wherein at least one ring atom (e.g. 1, 2, 3 or 4 preferably 1 or 2) is heteroatom selected from the group consisting of N, O and S.
  • heterocycle is 4-8 membered ring, more preferably 5-6 membered ring.
  • Examples of 4 membered heterocycle comprise but not limited to azetidinyl.
  • Examples of 5 membered heterocycle comprise but not limited to pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidinyl.
  • Examples of 6 membered heterocycle comprise but not limited to piperidinyl, morpholinyl, piperazinyl.
  • Examples of 7 membered heterocycle comprise but not limited to azacycloheptanyl, or the like.
  • the term “pharmaceutical combination” refers to a combined form of two or more active agents. It will be understood that these agents may be in a mixed or integrated form, e.g. a composition or mixture; or in a separated form, for example in separated compartments of a kit or in different kits.
  • the agents in the pharmaceutical combination may be given in an administration schedule that is synchronous, serial, overlapping, alternating, parallel, or any other treatment schedule in which the various agents are administered as part of a single treatment regimen.
  • the active ingredient is exemplified as one or more agents according to the invention, for example a compound of formula (I) or a pharmaceutically acceptable salt thereof or one or more additional cancer therapeutic agents as mentioned herein.
  • pharmaceutical composition refers to an active agent (s) , which is optionally combined with one or more pharmaceutically acceptable components (for example, but not limited to carrier) .
  • the active ingredient is exemplified as one or more agents according to the invention, for example a compound of formula (I) or a pharmaceutically acceptable salt thereof or one or more additional cancer therapeutic agents.
  • the pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
  • pharmaceutically acceptable carrier refers to those carriers which have no significant irritation and do not impair the bioactivity and property of the active compound. This term may also be understood as inert substance which is administered with active ingredient and is beneficial to the administration thereof. Non-limiting examples include but not limited to any of the following substances which may optionally be approved by Food and Drug Administration for use in human or animal: glidant, sweetening agent, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersant, disintegrant, suspending agent, stabilizing agent, isotonic agent, solvent or emulsifying agent.
  • the carriers comprise calcium carbonate, calcium phosphate, various sugars and starches, cellulose derivative, gelatine, vegetable oil and polyethylene glycol or the like. Other information regarding the carriers may be found in Remington: The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams &Wilkins (2005) , of which the contents are incorporated herein by reference.
  • administration refers to a method that enables a compound, composition or combination to be delivered to a desired site of biological action.
  • Such methods comprise but not limited to oral, parenteral (including intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular injection or infusion) , local, rectal administration or the like.
  • the term “effective amount” refers to the amount of a medicament or agent or combination which is sufficient to achieve the desired effect.
  • the effective amount may be determined individually and depends on the age and general condition of the receptor as well as specific active substance.
  • the effective amount in specific case can be determined by a person skilled in the art through conventional test.
  • the effective amount also refers to those of each of the agents exerting synergic effect.
  • active ingredient refers to a chemical entity useful for treating or preventing target disorder, disease or condition.
  • pharmaceutically acceptable refers to the compound, material, composition, combination and/or dosage form, which are within the scope of reliable medical judgment, suitable for contact with human and animal tissues, without over toxicity, irritation, allergic reaction or other problems or complications and has acceptable benefit/risk ratio.
  • kit of parts in the sense that the combination partners can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners, i.e., simultaneously or at different time points.
  • the parts of the kit of parts can then, e.g., be administered simultaneously or at different time points and with equal or different time intervals for any part of the kit of parts.
  • cancer and “cancerous tumor” have the same meaning herein, and include but not limited to solid tumors and blood cancers.
  • Exemplary solid tumors include but not limited to tumors of the breast, glioblastoma, bone, prostate, lung, adrenal gland (e.g., adrenocortical tumors) , bile duct, bladder, bronchus, nervous tissue (including neuronal and glial tumors) , gall bladder, stomach, salivary gland, esophagus, small intestine, cervix, colon, rectum, liver, ovary, pancreas, pituitary adenomas, and secretory adenomas.
  • Exemplary blood cancers include but not limited to lymphomas and leukemia.
  • Exemplary lymphomas include but not limited to multiple myeloma, Hodgkin's lymphoma, non-Hodgkin’s lymphomas (e.g., cutaneous T cell lymphomas such as Sezary syndrome and Mycosis fungoides, diffuse large cell lymphoma, HTLV-1 associated T cell lymphoma, nodal peripheral T cell lymphoma, extranodal peripheral T cell lymphoma, central nervous system lymphoma, and AIDS-related lymphoma) .
  • Exemplary leukemia include but not limited to acute and chronic types of both lymphocytic and myelogenous leukemia (e.g.
  • the “cancerous tumor” comprises triple negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma or multiple myeloma.
  • the “beneficial effect” herein refers to achieve additional advantageous therapeutic effects, diminish the incidence of side-effects or toxic effects (e.g., diarrhea or nausea) , delay or slow down progression of cancer, reduce the tumor volume in a cancer patient, prolong survival of a cancer patient, prevent or delay tumor metastasis, decrease mortality and morbidity; or to sensitize a cancer patient to the cancer therapeutic agent (s) when an IRE-1 ⁇ inhibitor is combined with the cancer therapeutic agent (s) ; or to reduce resistance to the cancer therapeutic agent (s) in a cancer patient who has been primarily resistant to such cancer therapeutic agent (s) .
  • the beneficial effect refers to exerting synergic effect as compared with either of combination partners employed alone.
  • subject or “patient” used herein refers to mammal subject or patient, preferably human subject or patient.
  • the present inventor surprisingly found that at least one beneficial effect for treating cancer is observed when the IRE1 ⁇ inhibitor as a compound of formula (I) or a pharmaceutically acceptable salt thereof is employed in combination therapy, for example one or more additional cancer therapeutic agents as recited herein.
  • a pharmaceutical combination comprising (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof and (b) one or more additional cancer therapeutic agents
  • R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1 -C 6 hydrocarbon chain containing 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
  • R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino
  • R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6 alkoxyl, C 1-6 hydroxylalkyl, C 1-6 alkoxylC 1-6 alkyl, and
  • R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6 alkoxyl C 1-6 alkyl; or
  • R 9 and R 10 together with the nitrogen atom to which they are attached form 3-10 membered heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6 alkylamino, C 1-6 alkoxyl.
  • the compound of formula (I) has the following formula (II) :
  • the additional cancer therapeutic agent (s) used in the combination according to the invention refers to a therapeutic agent (s) beyond the compound of formula (I) or pharmaceutically acceptable salt thereof as IRE1 ⁇ inhibitor.
  • Inositol requiring enzyme-1 ⁇ is a transmembrane stress-sensing and signaling molecule that controls the Unfolded Protein Response (UPR) .
  • URR Unfolded Protein Response
  • Numerous perturbations of protein folding contribute to Endoplasmic Reticulum (ER) stress.
  • ER Endoplasmic Reticulum
  • Downstream enzymatic activity is selectively activated during times of cellular stress, primarily during disease states and thus, inhibition of this pathway may impact tumor growth.
  • X-box protein 1 (XBP1) is activated in certain cancer types and may modulate the progression of disease. In vitro data shows that depletion of XBP1 inhibits tumor growth and relapse.
  • XBP1 splicing activation is up-regulated in cancer and increased following chemotherapy and therefore, is suspected to play a key role in drug resistance.
  • the present inventor surprisingly found that various types of physiological stress induce the unfolded protein response including but not limited to hypoxia, nutrient starvation, acidosis, and genetic damage resulting in mutant or over-expressed misfolded proteins (oncogenic stress) and one or more of these conditions are manifest in cancer cells, which may in part be mediated by the microenvironment of the tumor.
  • cytoprotective arm of the unfolded protein response UPR
  • bio-and chemotherapeutic drugs and radiation treatments may further impact the protein folding and degradation cycle in the ER thereby inducing the UPR as a protective resistance mechanism.
  • Patients succumb to cancer because either the tumor is resistant to conventional therapies or returns in a resistant form after an initial response to treatment.
  • IRE1 ⁇ inhibitor per se
  • the efficacy for treating cancer can be enhanced.
  • Chemotherapeutic agents, targeted small molecule oncology compounds, biomolecule etc. can directly induce ER stress and resulting UPR.
  • IRE1 ⁇ inhibitors can suppress this activation and thus can act synergistically for cellular proliferation inhibition when used in combination.
  • the additional cancer therapeutic agent (s) has at least one of the following features:
  • the one or more additional cancer therapeutic agents are selected from the group consisting of: cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents, e.g.
  • VEGF vascular endothelial growth factor
  • FGF/FGFR fibroblast growth factor
  • EGFR epidermal growth factor receptor
  • mTOR inhibitors corticosteroids
  • growth factor signal transduction kinase inhibitors mitochondrial dysfunction inducers
  • caspase activators chromatin disruptors and DNA repair enzyme inhibitors
  • HDAC inhibitors Bcr-Abl inhibitors
  • cancer therapeutic agents are as follows:
  • cytotoxic chemotherapeutic agents including microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) ;
  • antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) ;
  • pyrimidine analogs e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine
  • purine analogs e.g., folate antagonists and related inhibitors
  • folate antagonists and related inhibitors e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine
  • folic acid analogs e.g., methotrexate
  • antimitotic agents such as vinca alkaloids (e.g., eribulin, vinblastine, vincristine, and vinorelbine) ;
  • alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil) , ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa) , alkyl sulfonates-busulfan, nitrosoureas (e.g., carmustine (BCNU) and analogs, streptozocin) , and trazenes-dacarbazinine (DTIC) ;
  • nitrogen mustards e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil
  • ethylenimines and methylmelamines e.g., hexamethylmelamine and thiotepa
  • alkyl sulfonates-busulfan e.g., nitrosoureas
  • DNA damaging agents such as amsacrine, busulfan, camptothecin, irinotecan (CPT-11) , topotecan, chlorambucil, cyclophosphamide, cytoxan, hexamethylmelamineoxaliplatin, iphosphamide, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, teniposide, triethylenethiophosphoramide and etoposide (VP 16) ;
  • DNA damaging agents such as amsacrine, busulfan, camptothecin, irinotecan (CPT-11) , topotecan, chlorambucil, cyclophosphamide, cytoxan, hexamethylmelamineoxaliplatin, iphosphamide, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin
  • antitumor antibiotics such as actinomycin, dactinomycin (actinomycin D) , daunorubicin, doxorubicin (adriamycin) , epirubicin, idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin;
  • platinum coordination complexes such as cisplatin, carboplatin, and oxaliplatin
  • proteasome inhibitors including bortezomib ( [ (1R) -3-methyl-1- [ [ (2S) -1-oxo-3-phenyl-2-[ (pyrazinylcarbonyl) amino] propyl] amino] butyl] boronic acid; MG-341; MG-132 (N- [ (phenylmethoxy) carbonyl] -L-leucyl-N- [ (1 S) -1-formyl-3-methylbutyl] -L-leucinamide) , carfilzomib and ixazomib
  • HSP90 inhibitors including geldanamycin, radicicol, 17AAG, and Gamitrinib;
  • hormones and hormone analogs including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, apeledoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) ;
  • aromatase inhibitors e.g. letrozole, anastrozole
  • fibrinolytic agents such as tissue plasminogen activator, streptokinase and urokinase
  • fibrinolytic agents including aspirin, COX-2 inhibitors, dipyridamole, ticlopidine, clopidogrel, abciximab;
  • antimigratory agents e.g. somatostatin, wortmannin and PD98059;
  • antisecretory agents e.g. brefeldin
  • immunosuppressives including cyclosporine, tacrolimus (FK-506) , sirolimus (rapamycin) , azathioprine, mycophenolate mofetil;
  • anti-angiogenic compounds e.g., TNP 470, genistein
  • VEGF vascular endothelial growth factor
  • ZD6474 sunitinib, vatalanib, sorafenib, bevacizumab
  • FGF/FGFR fibroblast growth factor
  • FGF/FGFR inhibitors such as BGJ398, AZD4547, dovitinib, lenvatinib, JNJ-42756493, GP369, BAY1187982;
  • epidermal growth factor receptor (EGFR) inhibitors such as afatinib, gefitinib, erlotinib;
  • antibodies including trastuzumab bevacizumab cetuximab rituximab
  • checkpoint inhibitors including CTLA4 inhibitors
  • PD-1 inhibitors or PD-L1 inhibitors such as nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, in particular an antibody or binding fragment thereof;
  • cell cycle inhibitors and differentiation inducers e.g. tretinoin, ribociclib, palbociclib;
  • mTOR inhibitors including rapamycin, everolimus, sirolimus, temsirolimus, ridaforolimus; 24) corticosteroids, including cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone;
  • growth factor signal transduction kinase inhibitors such as imatinib, erlotinib, sorafenib, sunitinib, lapatinib, trametinib, temozolomide;
  • mitochondrial dysfunction inducers such as ⁇ -tocopherol, Bcl-2 and Bcl-XL inhibitors such as venetoclax, ABT-737, navitoclax, obatoclax mesylate;
  • caspase activators such as 25-hydroxycholesterol, mitomycin C, proscillaridin A, zearalenone, fumonisin B1, garcinol;
  • chromatin disruptors and DNA repair enzyme inhibitors including PARP inhibitors such as 3-aminobenzamide, olaparib, talazoparib, niraparib, veliparib, rucaparib;
  • HDAC inhibitors e.g. 17-AAG suberoylanilide hydroxamic acid
  • Bcr-Abl inhibitors including imatinib, nilotinib, dasatinib, bosutinib, ponatinib;
  • FMS-like tyrosine kinase 3 (Flt3) inhibitors including gilteritinib, lestaurtinib, midostaurin, nintedanib.
  • the additional cancer therapeutic agent is selected from the group consisting of:
  • Bcr-Abl inhibitors such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib, particularly nilotinib, dasatinib;
  • FMS-like tyrosine kinase 3 (Flt3) inhibitors such as gilteritinib, lestaurtinib, midostaurin, nintedanib, particularly lestaurtinib;
  • VEGF vascular endothelial growth factor
  • epidermal growth factor receptor (EGFR) inhibitors such as afatinib, gefitinib, erlotinib, particularly gefitinib;
  • mTOR inhibitors such as apamycin, everolimus, sirolimus, temsirolimus, ridaforolimus, particularly temsirolimus ;
  • HDAC inhibitors such as 17-AAG vorinostat particularly vorinostat ;
  • cytotoxic chemotherapeutic agents such as microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) , particularly taxane (e.g. paclitaxel, docetaxel) ;
  • antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) , particularly gemcitabine;
  • pyrimidine analogs e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine
  • purine analogs e.g., folate antagonists and related inhibitors
  • folate antagonists and related inhibitors e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine
  • folic acid analogs e.g., methotrexate
  • proteasome inhibitors such as bortezomib, MG-132, carfilzomib, ixazomib, particularly bortezomib;
  • hormones and hormone analogs including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, apeledoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) , particularly tamoxifen.
  • estrogen receptor inhibitors e.g. raloxifene, tamoxifen, apeledoxifene
  • androgen receptor inhibitors e.g. bicalutamide, nilutamide, enzalutamide
  • the additional cancer therapeutic agent is selected from the group consisting of cytotoxic chemotherapeutic agent, proteasome inhibitor, hormone analogue or any combination thereof. More preferably, the cytotoxic chemotherapeutic agent is selected from the group consisting of microtubule disruptors, e.g. paclitaxel or docetaxel. More preferably, the proteasome inhibitor is bortezomib. More preferably, the hormone analogue is anti-estrogen agent, e.g. tamoxifen.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and one cytotoxic chemotherapeutic agent.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor selected from paclitaxel or docetaxel.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and paclitaxel.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and a proteasome inhibitor.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and bortezomib.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and a hormone analogue.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and an estrogen receptor inhibitor.
  • a pharmaceutical combination comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and tamoxifen.
  • agents in the pharmaceutical combination according to the invention encompass their other forms like stereoisomers, salts, prodrugs as well as crystal modifications, e.g. solvates and polymorphs and such forms are within the scope of the present invention.
  • these forms are pharmaceutically acceptable.
  • the effective amounts or dosages of the compound of formula (I) or the pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents employed in the pharmaceutical combination according to the invention may vary depending on the particular compound or agent (s) employed, the mode of administration, the condition being treated, and severity of the condition being treated etc.
  • the dosage regimen is selected in accordance with a variety of factors including the route of administration, the renal and hepatic function of the subject or the like.
  • a physician, clinician or veterinarian of ordinary skill can readily determine and prescribe the effective amount required to prevent, counter or arrest the progress of the condition.
  • the effective dosage of the compound of formula (I) or the pharmaceutically acceptable salt thereof for daily use is about 10-2000 mg, preferably 50-1000 mg for a warm-blooded animal like human of about 70 kg bodyweight.
  • the effective dosage of the one or more additional cancer therapeutic agents for daily use in a warm-blooded animal, including man, can be determined by a package insert when said agent is provided as a marketed drug. It might be also be possible that the effective dosage of the one or more additional cancer therapeutic agents is adjusted according to species, age, individual condition, mode of administration, the clinical picture in question, etc.
  • the pharmaceutical combination of the invention comprises
  • the additional cancer therapeutic agents are defined as above.
  • the pharmaceutical combination according to the invention can further comprise one or more pharmaceutically acceptable carriers.
  • the pharmaceutical combination is provided in a unique form such as a pharmaceutical composition or mixture, the compound or agent (s) contained therein are combined with the same pharmaceutically acceptable carriers, for simultaneous, separate or sequential use.
  • composition according to the invention can be prepared in a manner known per se and are those suitable for enteral, such as oral or rectal, and parenteral administration to mammals (warm-blooded animals) , including man, comprising a therapeutically effective amount of compound of formula (I) and at least a therapeutically effective amount of cancer therapeutic agent, or further in combination with one or more pharmaceutically acceptable carries, especially suitable for enteral or parenteral application.
  • the agents contained therein, either the compound of formula (I) or the additional cancer therapeutic agent (s) are independently combined with the pharmaceutically acceptable carriers.
  • the pharmaceutically acceptable carriers for each of the agent may be identical or different according to practice requirement. Accordingly, provided is also a kit, comprising (a) a compound of formula (I) or pharmaceutically acceptable salt thereof and optional one or more pharmaceutically acceptable carriers; (b) one or more additional cancer therapeutic agents and optional one or more pharmaceutically acceptable carriers; and (c) instruction for using (a) and (b) .
  • the compound of formula (I) or pharmaceutically acceptable salt thereof and the additional cancer therapeutic agents are defined as above.
  • the ratio of the total amounts of the compound of formula (I) or the pharmaceutically acceptable salt thereof to one or more additional cancer therapeutic agents in the pharmaceutical combination according to the invention can be varied, e.g. in order to cope with the needs of a patient sub-population to be treated or the needs of a single patient which different needs can be due to the particular disease, age, sex, body weight, etc.
  • a method for treating cancerous tumor comprising administering a subject in need thereof an effective amount of the pharmaceutical combination according to the invention, wherein the active agents comprised in the pharmaceutical combination are defined as above.
  • the pharmaceutical combination comprises (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof and (b) one or more additional cancer therapeutic agents:
  • R 3 , R 4 , R 5 and R 6 are defined as above.
  • a method for treating cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents:
  • R 3 , R 4 , R 5 and R 6 are defined as above.
  • the one or more additional cancer therapeutic agents are defined as above.
  • the cancer therapeutic agent has at least one of the following features: (1) inducing ER stress; (2) inducing or up-regulating IRE-1 ⁇ expression; (3) inducing or up-regulating XBP1 splicing; and (4) being less effective when IRE-1 ⁇ is expressed.
  • said treatment is to cure the disease or to have an effect on disease regression or on the delay of progression of the disease.
  • said treatment is to inhibit the growth of tumor, for example, to reduce tumor volume, to delay the growth of tumor, to reverse the growth of tumor or any combination thereof.
  • said treatment is to kill the tumor, for example, to maintain the growth under a very low level.
  • the agents comprised therein are intended for simultaneous, separate or sequential use.
  • the agents comprised therein are intended for simultaneous, separate or sequential use.
  • such agents are intended for simultaneous, separate or sequential use.
  • the compound of formula (I) or pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents can be used, e.g. as a combined preparation or a pharmaceutical composition/mixture such that they can be administered at essentially the same time.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents can be in different compartments of a kit or different kits such that they can be administered at different time.
  • the compound of formula (I) or the pharmaceutically acceptable salt thereof can be administered before, after or along with the additional cancer therapeutic agent. The time interval between administrations of these agents may be several minutes, hours, days, months or even longer according to practical requirement.
  • the time intervals are such that the effect on the treated cancer in the combined use is larger than the effect which would be obtained by use of only any one of the combination partners.
  • the agents in the pharmaceutical combination according to the invention may be administered in the same or different routes.
  • the compound of formula (I) or pharmaceutically acceptable salt thereof and the additional cancer therapeutic agent (s) may be both administered orally or intravenously.
  • the compound of formula (I) or pharmaceutically acceptable salt thereof may be administered orally while the additional cancer therapeutic agent (s) may be administered intravenously and vice versa. It would be understood that when more than one additional cancer therapeutic agents are used, their administration routes are selected independently, i.e. either identical or different.
  • the compound of formula (I) has the following formula (II) :
  • a method for enhancing the efficacy of a cancer therapeutic agent comprising applying a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with the cancer therapeutic agent;
  • R 3 , R 4 , R 5 and R 6 are defined as above.
  • the compound of formula (I) has the following formula (II) :
  • the cancer therapeutic agent has at least one of the following features:
  • the enhancement of efficacy is embodied in inhibiting the growth of tumor, for example, reducing tumor volume, delaying the growth of tumor, reversing the growth of tumor or any combination thereof.
  • enhancement of efficacy is embodied in killing the tumor, for example, maintaining the growth under a very low level.
  • the one or more additional cancer therapeutic agents or the cancer therapeutic agent of which the efficacy to be enhanced are selected from the group consisting of cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents, e.g.
  • VEGF vascular endothelial growth factor
  • FGF/FGFR fibroblast growth factor
  • EGFR epidermal growth factor receptor
  • mTOR inhibitors corticosteroids
  • growth factor signal transduction kinase inhibitors mitochondrial dysfunction inducers
  • caspase activators chromatin disruptors and DNA repair enzyme inhibitors
  • HDAC inhibitors Bcr-Abl inhibitors
  • cancer therapeutic agents are as follows:
  • cytotoxic chemotherapeutic agents including microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) ;
  • antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) ;
  • pyrimidine analogs e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine
  • purine analogs e.g., folate antagonists and related inhibitors
  • folate antagonists and related inhibitors e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine
  • folic acid analogs e.g., methotrexate
  • antimitotic agents such as vinca alkaloids (e.g., eribulin, vinblastine, vincristine, and vinorelbine) ;
  • alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil) , ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa) , alkyl sulfonates-busulfan, nitrosoureas (e.g., carmustine (BCNU) and analogs, streptozocin) , and trazenes-dacarbazinine (DTIC) ;
  • nitrogen mustards e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil
  • ethylenimines and methylmelamines e.g., hexamethylmelamine and thiotepa
  • alkyl sulfonates-busulfan e.g., nitrosoureas
  • DNA damaging agents such as amsacrine, busulfan, camptothecin, irinotecan (CPT-11) , topotecan, chlorambucil, cyclophosphamide, cytoxan, hexamethylmelamineoxaliplatin, iphosphamide, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, teniposide, triethylenethiophosphoramide and etoposide (VP 16) ;
  • DNA damaging agents such as amsacrine, busulfan, camptothecin, irinotecan (CPT-11) , topotecan, chlorambucil, cyclophosphamide, cytoxan, hexamethylmelamineoxaliplatin, iphosphamide, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin
  • antitumor antibiotics such as actinomycin, dactinomycin (actinomycin D) , daunorubicin, doxorubicin (adriamycin) , epirubicin, idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin;
  • platinum coordination complexes such as cisplatin, carboplatin, and oxaliplatin
  • proteasome inhibitors including bortezomib ( [ (1R) -3-methyl-1- [ [ (2S) -1-oxo-3-phenyl-2- [ (pyrazinylcarbonyl) amino] propyl] amino] butyl] boronic acid; MG-341; ) , MG-132 (N- [ (phenylmethoxy) carbonyl] -L-leucyl-N- [ (1 S) -1-formyl-3-methylbutyl] -L-leucinamide) , carfilzomib and ixazomib
  • HSP90 inhibitors including geldanamycin, radicicol, 17AAG, and gamitrinib;
  • hormones and hormone analogs including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, apeledoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) ;
  • aromatase inhibitors e.g. letrozole, anastrozole
  • fibrinolytic agents such as tissue plasminogen activator, streptokinase and urokinase
  • fibrinolytic agents including aspirin, COX-2 inhibitors, dipyridamole, ticlopidine, clopidogrel, abciximab;
  • antimigratory agents e.g. somatostatin, wortmannin and PD98059;
  • antisecretory agents e.g. brefeldin
  • immunosuppressives including cyclosporine, tacrolimus (FK-506) , sirolimus (rapamycin) , azathioprine, mycophenolate mofetil;
  • anti-angiogenic compounds e.g., TNP 470, genistein
  • VEGF vascular endothelial growth factor
  • ZD6474 sunitinib, vatalanib, sorafenib, bevacizumab
  • FGF/FGFR fibroblast growth factor receptor
  • epidermal growth factor (EGFR) inhibitors such as afatinib, gefitinib, erlotinib;
  • antibodies including trastuzumab bevacizumab cetuximab rituximab
  • checkpoint inhibitors including CTLA4 inhibitors
  • PD-1 inhibitors or PD-L1 inhibitors such as nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, in particular an antibody or binding fragment thereof;
  • cell cycle inhibitors and differentiation inducers e.g. tretinoin, ribociclib, palbociclib;
  • mTOR inhibitors including rapamycin, everolimus, sirolimus, temsirolimus, ridaforolimus;
  • corticosteroids including cortisone, dexamethasone, hydrocortisone, methylpednisolone,prednisone, and prenisolone;
  • growth factor signal transduction kinase inhibitors such as imatinib, erlotinib, sorafenib, sunitinib, lapatinib, trametinib, temozolomide;
  • mitochondrial dysfunction inducers such as ⁇ -tocopherol, Bcl-2 and Bcl-XL inhibitors such as venetoclax, ABT-737, navitoclax, obatoclax mesylate;
  • caspase activators such as 25-hydroxycholesterol, mitomycin C, proscillaridin A, zearalenone, fumonisin B1, garcinol;
  • chromatin disruptors and DNA repair enzyme inhibitors including PARP inhibitors such as 3-aminobenzamide, olaparib, talazoparib, niraparib, veliparib, rucaparib;
  • HDAC inhibitors e.g. 17-AAG suberoylanilide hydroxamic acid
  • Bcr-Abl inhibitors including imatinib, nilotinib, dasatinib, bosutinib, ponatinib;
  • FMS-like tyrosine kinase 3 (Flt3) inhibitors including gilteritinib, lestaurtinib, midostaurin, nintedanib.
  • the additional cancer therapeutic agent is selected from the group consisting of:
  • Bcr-Abl inhibitors such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib, particularly nilotinib, dasatinib;
  • FMS-like tyrosine kinase 3 (Flt3) inhibitors such as gilteritinib, lestaurtinib, midostaurin, nintedanib, particularly lestaurtinib;
  • VEGF vascular endothelial growth factor
  • epidermal growth factor receptor (EGFR) inhibitors such as afatinib, gefitinib, erlotinib, particularly gefitinib;
  • mTOR inhibitors such as apamycin, everolimus, sirolimus, temsirolimus, ridaforolimus, particularly temsirolimus ;
  • HDAC inhibitors such as 17-AAG vorinostat particularly vorinostat ;
  • cytotoxic chemotherapeutic agents such as microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) , particularly taxane (paclitaxel, docetaxel) ;
  • antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) , particularly gemcitabine;
  • pyrimidine analogs e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine
  • purine analogs e.g., folate antagonists and related inhibitors
  • folate antagonists and related inhibitors e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine
  • folic acid analogs e.g., methotrexate
  • proteasome inhibitors such as bortezomib, MG-132, carfilzomib, ixazomib, particularly bortezomib;
  • hormones and hormone analogs including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, apeledoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) , particularly tamoxifen.
  • estrogen receptor inhibitors e.g. raloxifene, tamoxifen, apeledoxifene
  • androgen receptor inhibitors e.g. bicalutamide, nilutamide, enzalutamide
  • the additional cancer therapeutic agent used in the method for treatment of cancerous tumor or the cancer therapeutic agent of which the efficacy to be enhanced is selected from the group consisting of cytotoxic chemotherapeutic agent, proteasome inhibitor, hormone analogue or any combination thereof. More preferably, the cytotoxic chemotherapeutic agent is selected from the group consisting of microtubule disruptors, e.g. paclitaxel or docetaxel. More preferably, the proteasome inhibitor is bortezomib. More preferably, the hormone analogue is anti-estrogen agent, e.g. tamoxifen.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and one cytotoxic chemotherapeutic agent.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor selected from paclitaxel or docetaxel.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and paclitaxel.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and a proteasome inhibitor.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and bortezomib.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and a hormone analogue.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and an estrogen receptor inhibitor.
  • a method for treatment of cancerous tumor comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and tamoxifen.
  • the pharmaceutical combination comprises (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof and (b) one or more additional cancer therapeutic agents:
  • R 3 , R 4 , R 5 and R 6 are defined as above.
  • the compound of formula (I) has the following formula (II) :
  • the additional cancer therapeutic agents are defined as above.
  • the cancerous tumors which can be treated with the pharmaceutical combination according to the invention or to which the efficacy of the cancer therapeutic agent can be enhanced comprise but not limited to solid tumors and blood cancers.
  • Exemplary solid tumors include, but are not limited to tumors of breast, glioblastoma, bone, prostate, lung, adrenal gland (e.g., adrenocortical tumors) , bile duct, bladder, bronchus, nervous tissue (including neuronal and glial tumors) , gall bladder, stomach, salivary gland, esophagus, small intestine, cervix, colon, rectum, liver, ovary, pancreas, pituitary adenomas, and secretory adenomas.
  • Blood cancers include lymphomas and leukemia.
  • Exemplary lymphomas include, but are not limited to multiple myeloma, Hodgkin's lymphoma, non-Hodgkin’s lymphomas (e.g., cutaneous T cell lymphomas such as Sezary syndrome and Mycosis fungoides, diffuse large cell lymphoma, HTLV-1 associated T cell lymphoma, nodal peripheral T cell lymphoma, extranodal peripheral T cell lymphoma, central nervous system lymphoma, and AIDS-related lymphoma) .
  • Exemplary leukemia include, but are not limited to acute and chronic types of both lymphocytic and myelogenous leukemia (e.g.
  • acute lymphocytic or lymphoblastic leukemia acute myelogenous leukemia, acute myeloid leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T cell prolymphocyte leukemia, adult T cell leukemia, and hairy cell leukemia.
  • the cancer is selected from liver cancer, breast cancer, lung cancer, ovarian cancer, head and neck cancer, glioblastoma, and multiple myeloma.
  • the breast cancer is triple negative breast cancer. More preferably, triple negative breast cancer is metastatic, recurrent, refractory or advanced.
  • the breast cancer is estrogen positive breast cancer. More preferably, estrogen positive breast cancer is metastatic, recurrent, refractory or advanced.
  • the lung cancer is non-small cell lung carcinoma (NSCLC) . More preferably, non-small cell lung carcinoma is metastatic, recurrent, refractory or advanced.
  • NSCLC non-small cell lung carcinoma
  • the lung cancer is small cell lung carcinoma (SCLC) . More preferably, small cell lung carcinoma is metastatic, recurrent, refractory or advanced.
  • SCLC small cell lung carcinoma
  • the ovarian cancer is metastatic, recurrent, refractory or advanced.
  • the head and neck cancer is metastatic, recurrent, refractory or advanced.
  • glioblastoma is metastatic, recurrent, refractory or advanced.
  • multiple myeloma is metastatic, recurrent, refractory or advanced.
  • the present inventor has performed a 7-day toxicity study based on cynomolgus monkey model showed that Orin1001 as the compound of formula (I) or pharmaceutically acceptable salt thereof has a No Observable Aderse Effect Level (NOAEL) of 150 mg/kg/d, and a 5-fold safety margin amounting to 750 mg/kg/d, which indicates a good safety profile of such compound and thus it can be used in a relatively high dosage without significant side effect like toxicity, either alone or in combination with other cancer therapeutical agent.
  • NOAEL No Observable Aderse Effect Level
  • the pharmaceutical combination according to the invention can be used to effectively treat cancer/tumor by inhibiting tumor growth or killing tumor, for example delaying, arresting, or reversing tumor growth with synergist effect and good safety.
  • PO oral; sc: subcutaneous; iv: intravenous; qod: every other day; qwk: once a week; qd: once a day.
  • IRE1 can be activated indirectly by a number of small molecules.
  • MM. 1S CRL-2974 TM
  • HEK-293 CRL-1573 TM
  • H929 CRL-9068 TM
  • RPMI8226 CCL-155 TM
  • A549 CCL-185 TM
  • HT-29 HTB-38 TM
  • MCF7 HTB-22 TM
  • Hep G2 Hep G2 ( HB-8065 TM ) is “Hepatoma” , human hepatocellular carcinoma.
  • This method applies to any mammalian cell line but was typically applied to human MM1s myeloma cells for EC 50 and RPMI8826 plasmacytoma cells for confirmation of selected compounds. Briefly, cells were grown in standard conditions and spread into 96 well tissue culture plates. Cells were treated with compounds with indicated concentration using serial dilutions. DTT (dithiothreitol) or compounds were added at the same time and cells were harvested after indicated hour’s treatment. Cells treated with DTT alone were used as 100% XBP1s positive controls and cell left untreated were used as base line XBP1s level.
  • Examples 1-12 various compounds were tested for their respective induction to cells lines’ ER stress measured by XBP1s level and the results are shown in Figures 1-12, respectively.
  • Example 1 showed that Lestaurtinib enhanced multiple cell lines’ ER stress measured by XBP1s level (Figure 1) .
  • Example 2 showed that Nilotinib enhanced MM1c cell’s ER stress measured by XBP1s level ( Figure 2) . The same methods were used as above for Lestaurtinib.
  • Example 3 showed that Sorafenib enhanced A549 cell lines’ ER stress measured by XBP1s level ( Figure 3) . The same methods were used as above for Lestaurtinib.
  • Example 4 showed that Dasatinib enhanced A549 cell lines’ ER stress measured by XBP1s level ( Figure 4) .
  • the same methods were used as above for Lestaurtinib.
  • Example 5 showed that Gefitinib enhanced A549 cell lines’ ER stress measured by XBP1s level ( Figure 5) . The same methods were used as above for Lestaurtinib.
  • Example 6 showed that Lestaurtinib, temisirolimus, vatalinib enhanced several cell lines’ ER stress measured by XBP1s level ( Figure 6) . The same methods were used as above for Lestaurtinib.
  • Example 7 showed that Torisel (Temsirolimus) enhanced A549 cell lines’ ER stress measured by XBP1s level ( Figure 7) . The same methods were used as above for Lestaurtinib.
  • Example 8 showed that Vorinostat enhanced HT-29 cell lines’ ER stress measured by XBP1s level ( Figure 8) . The same methods were used as above for Lestaurtinib.
  • Example 9 showed that Paclitaxel enhanced RPMI 8226 cell lines’ ER stress measured by XBP1s level ( Figure 9) . The same methods were used as above for Lestaurtinib.
  • Example 10 showed that Gemcitabine enhanced RPMI 8226 cell lines’ ER stress measured by XBP1s level ( Figure 10) . The same methods were used as above for Lestaurtinib.
  • Example 11 showed that 17-AAG enhanced MCF-7 cell lines’ ER stress measured by XBP1s level ( Figure 11) . The same methods were used as above for Lestaurtinib.
  • Example 12 showed that 17-AAG enhanced Hepatoma cell lines’ ER stress measured by XBP1s level ( Figure 12) . The same methods were used as above for Lestaurtinib.
  • Example 13 showed Intratumoral XBP-1 spliced effect of IRE-1 compound Orin1001 in treated RPMI xenografts (Figure 13) .
  • Figure 13 Nude mice with xenografts using RPMI 8226 tumor cells were treated by IV injection of at 0.8 mg/kg after the tumor established in 21 days. The mice were treated again on the 24 th day with On Day 27, the mice were treated with Orin1001 at 30 mg/kg PO, four hours later the mice were sacrificed as in Example 22, and tumor tissues were isolated.
  • RNA extraction and RT-PCR analysis gave the results as shown Figure 13.
  • Examples 14-22 were in vivo tests for efficacies of ORIN 1001 in combination with other cancer therapeutic agents and the procedures were summarized as follows.
  • IRE-1 ⁇ inhibitor in combination with a cytotoxic agent or hormone antagonist may be more effective in inhibiting tumor growth and prevent tumor relapse.
  • a novel, first-in-class IRE-1 ⁇ inhibitor, Orin1001 was evaluated in combination with paclitaxel, tamoxifen or in mouse tumor xenograft models, which were developed by Charles River Laboratories and the studies were performed by Charles River Laboratories as contracted services. These models include triple negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma and multiple myeloma. All the therapeutic agents such as paclitaxel, tamoxifen and were purchased by Charles River from commercial sources.
  • Example 14 Orin1001 inhibits triple negative breast cancer in combination with paclitaxel at different tumor growth stage
  • Orin1001 was administered by oral gavage in combination with paclitaxel in a xenograft mouse model using female NCr nu/nu mice injected subcutaneously with human breast adenocarcinoma MDA-MB231 tumor cells.
  • dosing was initiated either on Day 1 (when tumors reached 225-250 mm 3 ) , Day 14 or Day 28 of tumor growth.
  • Orin1001 In combination with paclitaxel, Orin1001 showed significant tumor inhibition compared to paclitaxel alone at all stages of tumor growth. Specifically, Orin1001 (300mg/kg) was applied at Day 1, Day 14 and Day 28 respectively in combination with paclitaxel (10 mg/kg) showing the effects of intervene of Orin1001 at any stage of tumor growth.
  • Orin1001 and paclitaxel could delay the growth of tumor and the synergist effect could be seen at every intervening stage like early, middle or late stage, even starting as late as Day 28.
  • the growth of tumor could be reversed when Orin1001 is applied together with paclitaxel at every intervening stage, even starting as late as Day 28.
  • extended oral dosing of Orin1001 for up to 60 consecutive days was well tolerated and also resulted in a significant synergist effect on tumor inhibition.
  • Example 15 Orin1001 inhibits triple negative breast cancer in combination with Paclitaxel dose proportionally
  • Orin1001 showed significant tumor inhibition compared to paclitaxel alone at all stages of tumor growth.
  • Treatment with 300 mg/kg Orin1001 in combination with paclitaxel resulted in 3 partial regressions and 1 tumor-free survival versus 1 partial regression in the paclitaxel group alone.
  • Orin1001 was applied at a dose at 150 mg/kg/day or more, the inhibitory effects were much obvious.
  • the growth of tumors could be reversed.
  • Orin1001 was applied at a dose of 300 mg/kg, the tumor growth was almost arrested.
  • extended oral dosing of Orin1001 for up to 60 consecutive days was well tolerated and resulted in a significant synergist effect on tumor inhibition.
  • Example 16 Orin1001 inhibits estrogen positive breast cancer in combination with tamoxifen
  • Orin1001 in combination with tamoxifen showed significant tumor inhibition compared to tamoxifen alone.
  • Example 17 Orin1001 inhibits estrogen positive breast cancer in combination with paclitaxel
  • Orin1001 in combination with paclitaxel showed significant synergistic effects on tumor growth inhibition compared to paclitaxel alone. Specifically, when Orin1001 was applied in combination with paclitaxel, a synergistic effect could be seen over Orin1001 or paclitaxel alone and particularly the growth of tumor was almost arrested and then reversed under the combined use.
  • Example 18 Orin1001 inhibits ovarian cancer in combination with paclitaxel
  • TGD (%) [T-C/C] x 100, where T-C is the difference in time to tumor endpoint from Treated (T) and Control (C).
  • TGD was 29, 40 and 68% for Orin1001 alone, paclitaxel alone and Orin1001 in combination with paclitaxel, respectively.
  • Figure 18 it can be seen that in combination with Paclitaxel, 300 mg/kg Orin1001 showed increased tumor inhibition compared to paclitaxel alone and the reverse of tumor growth was observed under the combined use of Orin1001 and paclitaxel.
  • Example 19 Orin1001 inhibits glioblastoma in combination with paclitaxel
  • TGD (%) [T-C/C] x100; where T-C is the difference in time to tumor endpoint from Treated (T) and Control (C).
  • TGD was 13, 17 and 50% for Orin1001 alone, paclitaxel alone and Orin1001 in combination with paclitaxel, respectively.
  • TTE time to tumor endpoint
  • Tumor growth delay and survival were significantly greater with Orin1001 in combination with paclitaxel than with paclitaxel alone (p ⁇ 0.01, Chi Square and Gehan-Breslow-Wilcoxon test) .
  • 300 mg/kg Orin1001 showed a marked increase in tumor inhibition compared to paclitaxel alone with 2 animals showing partial regression versus 0 animals in the other treated groups.
  • the tumor delay effects was not very significant for Orin1001 or paclitaxel each alone over the control and the tumor volumes reach the maximum at about Day 30.On the contrary, when they are in combined use, the tumor growth was significantly delayed and the effect of growth reverse was also observed as shown in Figure 19.
  • Example 20 Orin1001 inhibits non-small cell lung cancer in combination with paclitaxel
  • Example 21 Orin1001 inhibits liver cancer in combination with sorafenib
  • Example 22 Orin1001 was chosen to be used in combination with other therapeutic agents
  • Example 22 was related to comparative PD/PK data of Orin 1001 VS compound 4315, as its structure shown below, which is an earlier lead of this series of compounds. Tunicamycin was used herein to activate IRE1, then Orin1001 (also called 4485) or 4315 were given to inhibit the activating effect. Orin1001 demonstrated potent in vivo potency inhibiting IRE1 ⁇ in our liver PD screening assay. The results are shown in Figure 22. Each gel panel represents one mouse liver sample as in the figure.
  • mice BALB/c mice was injected intraperitoneally with 100 microliters at an equivalent dose of 1 mg/kg tunicamycin solution. Two hours after tunicamycin injection, mice were dosed with compound of interest either PO or IV. Following 2 hours for PO delivery of compound, mice were euthanized according to IACUC protocols using CO 2 from a compressed air source. A 1 cm 3 fragment of the liver for homogenization and extraction of the RNA were collected for further analysis. Total RNA is harvested from cells or tissue using TRIzol according to the manufacturer’s procedures. After ethanol precipitation and resuspension of the RNA, RiboGreen (Invitrogen) is used to quantify the yield and normalize the RNA concentration in the source tube containing isolated RNA.
  • RiboGreen Invitrogen
  • RT-PCR is performed by Oligo (dT) priming, and SuperScript II (Invitrogen) transcription using the Amplitaq Gold Kit (Applied Biosystems) according to the manufacturer’s protocols.
  • Primers for human XBP-1 are 5_-CCTGGTTGCTGAAGAGGAGG-3_ (forward, Seq ID No. 8) and 5_-CCATGGGGAGATGTTCTGGAG-3_ (reverse, Seq ID No. 9)
  • Primers for mouse are 5_-ACACGCTTGGGAATGGACAC-3_ (forward, Seq ID No. 10) and 5_-CCATGGGAAGATGTTCTGGG-3 (Seq ID No. 11) . All DNA oligos were purchased from IDT DNA Technologies.
  • PCR is run on a Bio-Rad PTC-100 96-well thermocycler with heating at 94 °C for 30 s, annealing at 58 °C for 30 s, and polymerizing at 72 °C for 30 s for 35 cycles. Reactions are run on 4% precast NuSieve gels from Cambrex and visualized by ethidium bromide staining and UV excitation.
  • mXBP-1 572 5’ -AGATGTTCTGGGGAGGTGACAACT-3’ custom order, IDT
  • mGAPDH 548 custom order, IDT
  • PK experiments are standard tests that performed in either WuXi PharmaTech or Charels River Laboratory. 4315 and 4485 (Orin1001) were dosed PO as a suspnesion in 1% microcellulose (Sigma) and 50% Sucrouse (Sigma) .
  • Orin1001 has an ED 50 less than 2 mg/kg PO vs 4315 which has an ED 50 >10 mg/kg. 4315 ED 50 was determined to be 50 mg/kg in a separate experiment.
  • Compound 4315 is disclosed in WO2011/127070 A2 as compound B, a preferred IRE1 ⁇ inhibitor.
  • top panel when labeled as PBS/4315 or Tun/4315, they meant the mice was either dosed with PBS buffer or tunicamycin to active the IRE1 ⁇ so that XBP1s was observed as illustrated in the lower panel.
  • all the 4485 dosing group mice were dosed firstly with tunicamycin.
  • Orin1001 (4485) also has much improved oral bioavilability as shown in the table cross all tested spices.
  • oral bioavailability 4485 (Orin1001) oral bioavilability Mouse 23% (5mg/kg dose) 63% (10mg/kg dose) Rat 36% (10mg/kg dose) 52% (10mg/kg dose) Dog 17% (10mg/kg dose) 68.5% (4mg/kg dose)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided are a pharmaceutical combination comprising an IRE1α inhibitor and one or more additional cancer therapeutic agents for the treatment of cancerous tumor, a pharmaceutical composition containing the same and a method for treating cancerous tumor using the same.

Description

[Title established by the ISA under Rule 37.2] IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT TECHNICAL FIELD
Provided are a pharmaceutical combination comprising an IRE1α inhibitor and one or more additional cancer therapeutic agents for the treatment of cancerous tumor, a pharmaceutical composition containing the same and a method for treating cancerous tumor using the same.
BACKGROUND
Cancer or cancerous tumor is the second leading cause of death in the developed world and is expected to kill more than 500,000 people in the United States this year. Despite sophisticated early detection techniques, new therapies and improved outcomes, new treatments are still required to improve patients’ lives. One such area to this end is using combination therapies to target cancer from multiple weak points or multiple oncogenic drivers. Very often, cancer responds to treatments initially but the cancer reoccurs due to resistance and renewal of cancer stem cell survivors. Surgery, chemotherapy and radiotherapy, the traditional anti-cancer methods which may not result in complete responses or “cures” , can now be combined with targeted therapies and immunotherapies to improve patient survival outcomes versus using them as single agents. However, new and novel combinations are desirable to achieve greater therapeutic successes and improved patient outcomes.
The tumor microenvironment represents an underutilized therapeutic target area which impacts solid tumor growth and survival. Small molecule modulators of IRE-1α kinase and RNase functions have been reported with distinct mechanisms of action reflecting the engagement physically distinct binding sites and direct RNAse active site binding compounds represent a class of modulators that potently, reversibly, and selectively inhibit IRE-1α RNase activity including naphthalene (WO 2008/154484 A1; WO 2011/056744 A1) and coumarin (WO 2011/127070 A2) aromatic systems and which may be used as therapeutic agents to treat tumors. Although the IRE-1 inhibitor has been studied, as the effective cancer therapy is needed, it is desirable to have effective combined therapy of such inhibitor with other cancer therapeutic agent (s) to maximum the treatment efficacy against various tumors.
SUMMARY
In an aspect, provided is a pharmaceutical combination comprising
(a) a compound of formula (I) or a pharmaceutically acceptable salt thereof, and (b) one or more additional cancer therapeutic agents,
Figure PCTCN2018096613-appb-000001
wherein
R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1-C 6 hydrocarbon chain containing 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino;
R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6alkoxyl, C 1-6hydroxylalkyl, C 1-6alkoxylC 1-6alkyl,
Figure PCTCN2018096613-appb-000002
and 
Figure PCTCN2018096613-appb-000003
R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6alkoxyl C 1-6alkyl; or
R 9 and R 10 together with the nitrogen atom to which they are attached form 3-10 membered heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6alkylamino, C 1-6 alkoxyl.
In an embodiment of the invention, the pharmaceutical combination is provided in the form of a pharmaceutical composition. In an alternative embodiment of the invention, the pharmaceutical combination is provided in the form of one or more kits.
In a further aspect, provided is a method for treating cancerous tumor, comprising administering a subject in need thereof an effective amount of the pharmaceutical combination according to the invention.
In a yet further aspect, provided is a method for enhancing the efficacy of a cancer therapeutic agent comprising applying the compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with the cancer therapeutic agent.
In a specific embodiment of the invention, the compound of formula (I) has the following  formula (II) (which is also designated hereinafter as compound Orin1001 or compound 4485) :
Figure PCTCN2018096613-appb-000004
Brief Description of the Drawings
Figure 1: Induction of percentage of XBP1s relative to total XBP1s and XBP1u measured by RT-qPCR to increasing concentrations of Lestaurtinib (X axis) in MM1s (circles) , HEK293 (triangles) , RPMI 8226 (diamonds) and H929 (squares) cells. Plots were generated using Excel fit software.
Figure 2: Nilotinib induced greater than 50 % XBP1s after 2 hours of treatment (triangles) but modest amounts at 1 (circles) or 4 hours (squares) of treatment of MM1s cells.
Figure 3: Sorafenib induces highest levels of XBP1s after 1 (circles) or 2 (triangles) hours of treatment with modest levels after 4 hour (squares) of treatment of A549 cells.
Figure 4: Dasatinib induces highest levels of XBP1s after 1 hour (circles) of treatment.
Figure 5: Gefitinib induces highest levels of XBP1s after 2 hours of treatment (triangles) increasing at 1 hour (circles) .
Figure 6: Indicated drugs induced potent XBP1s after 1 hour (circles) , 2 hours (triangles) or 4 hours (squares) of treatment shown by IC50 curves for Hepatoma (Hep G2, top panel) MCF-7 (mid panel) and RPMI8226 cells (bottom panel) .
Figure 7: Torisel
Figure PCTCN2018096613-appb-000005
induces high levels of XBP1s after 1 hour (circles) and 4 hours (squares) but modest levels after 2 hours of treatment (triangles) of A549 cells at indicated concentrations.
Figure 8: Vorinostat induces high levels of XBP1s after 1 hour (circles) and little after 2 hours (triangles) or 4 hours (squares) of treatment of HT-29 cells at indicated concentrations.
Figure 9: Paclitaxel induces high levels of XBP1s after 1 hour (circles) and with modest levels after 4 hours (squares) or 2 hours of treatment (triangles) of RPMI 8226 cells at indicated concentrations.
Figure 10: Gemcitabine induces high levels of XBP1s after 4 hours (squares) but low levels after 1 hour (circles) or after 2 hours of treatment (triangles) of RPMI 8226 cells at indicated concentrations.
Figure 11: 17-AAG induces high levels of XBP1s after 1 hour (circles) with modest levels after 2 hours (triangles) and low levels after 4 hours (squares) of treatment or of MCF-7 cells at indicated concentrations.
Figure 12: 17-AAG induces high levels of XBP1s after 1 hour (circles) with modest levels after 2 hours (triangles) and low levels after 4 hours (squares) of treatment of Hepatoma cells at indicated concentrations.
Figure 13: Intratumoral XBP-1 spliced effect of IRE-1 compound Orin1001 in
Figure PCTCN2018096613-appb-000006
treated RPMI xenografts
Figures 14-21: Synergistic effects of compound ORIN 1001 with other cancer therapeutic agents.
Figure 22: XBP-1 splicing analysis of liver from compounds dosed PO.
Detailed Description
Definition
Unless stated otherwise, the terms and phrases used herein have the following meaning. A specific term or phrase shall not be considered as unclear or indefinite when it is not specifically defined. It should be understood according to the general meaning in the art. The trade name used herein refers to the corresponding product or the active ingredient.
Unless specifically defined otherwise, proportion (including percentage) or part is calculated based on weight herein.
When used with a numerical variable, the term “approximate” or “about” usually refers to the value of the variable and all the values of the variable within the experimental error (for example, within an average 95% confidence interval) or within ± 10% of the specified value, or a wider range.
The expression “comprise” or its synonyms “contain” , “include” , “have” or the like is open- ended, which does not exclude other unlisted elements, steps or ingredients. The expression “consist of” excludes any unlisted elements, steps or ingredients. The expression “substantially consist of” refers to specified elements, steps or ingredients within a given range, together with optional elements, steps or components which do not substantively affect the basic and novel feature of the claimed subject matter. It will be understood that the expression “comprise” encompasses the expressions “substantially consist of” and “consist of” .
The term “optional” or “optionally” means the event described subsequent thereto may or may not happen. This term encompasses the cases that the event may or may not happen.
The term “C m-n” or “m-n membered” used herein means that the moiety has m-n carbon atoms or m-n atoms. For example, “C 1-6alkyl” means said alkyl has 1-6 carbon atoms. Likewise, C 3-10 cycloalkyl means said cycloalkyl has 3-10 carbon atoms. It will be understood that, when the term C m-n is used in a group containing a moiety other than C-containing moiety, it refers to the carbon atom number in said C-containing moiety. For example, “C 1-6” in C 1-6hydroxylalkyl or C 1- 6alkylamino means that the alkyl therein has 1-6 carbon atoms. In case more than one C-containing moieties are present, they are defined independently, for example C 1-6alkoxylC 1-6alkyl. If only one C m-n is defined, it should apply to all C-containing moieties, for example, C 1- 6alkoxylalkyl means the alkoxyl and alkyl therein are each C 1-6 moiety.
It will be understood that the numerical range herein refers to each of the integers therein and any sub-range constituted by the integers. For example, “C 1-6” means said group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. Accordingly, “C 1-6alkyl” encompasses “C 2-5alkyl” , “C 1-5alkyl” , “C 2-6alkyl” as well as C 1alkyl, C 2alkyl, C 3alkyl, C 4alkyl, C 5alkyl, C 6alkyl or the like.
The term “substitution” means one or more hydrogen atoms on a given atom are replaced by substituent (s) , provided that the valence of the given atom is normal and the compound after substitution is stable.
The expression “one or more” or “at least one” refers to one, two, three, four, five, six, seven, eight, nine or more.
When any variable (e.g. R) occurs at the structure of a compound over one time, it is defined independently at each case. Therefore, for example, if a group is substituted by 0-2 R, the group may be optionally substituted by at most two R and R has independent option at each case.
Additionally, a combination of substituents and/or the variants thereof are allowed only if such a combination will result in a stable compound.
Unless stated otherwise, the term “hetero” means heteroatom or heteroatom radical (i.e. a radical containing heteroatom) , i.e. the atoms beyond carbon and hydrogen atoms or the radical containing such atoms. Preferably, the heteroatom is independently selected from the group consisting of O, N, S and the like. In an embodiment wherein two or more heteroatoms are involved, the two or more heteroatoms may be the same, or part or all of the two or more heteroatoms may be different.
The term “alkyl” , either used alone or in combination with other group (s) , refers to a linear or branched saturated aliphatic hydrocarbyl group composed of carbon and hydrogen atoms. Non-limiting examples of C 1-6alkyl comprise but not limited to methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl or the like.
The term “alkoxyl” , either used alone or in combination with other group (s) , refers to an “alkyl” which is connected to the rest of the molecule via “-O-” , wherein the “alkyl” is defined as above. Non-limiting examples of C 1-6alkoxyl comprise but not limited to methoxyl, ethoxyl, propoxy or the like.
The term “cycloalkyl” , either used alone or in combination with other group (s) , refers to saturated monocyclic or polycyclic hydrocarbyl group composed of carbon and hydrogen atoms. Cycloalkyl may contain 3-10, for example, 3-8, 3-7, 3-6, 3-5, 4-7, 4-6, or 3-4 carbon atoms or the like, or 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. Non-limiting examples of C 3-10 cycloalkyl comprise but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or the like. The cycloalkyl may further optionally contain one or more (preferably 1 or 2) heteroatoms independently selected from the group consisting of N, O, and S.
The term “heterocycle” or “heterocyclic” , either used alone or in combination with other group (s) , refers to a saturated or unsaturated monocyclic or polycyclic system group, wherein part of the ring atoms (e.g. 1, 2, 3 or 4) are heteroatoms independently selected from the group consisting of N, O and S, and rest of the ring atoms are C. For example, 3-10 membered heterocycle contains 3-10 ring atoms in the system, wherein at least one ring atom (e.g. 1, 2, 3 or 4 preferably 1 or 2) is heteroatom selected from the group consisting of N, O and S. Preferably, heterocycle is 4-8 membered ring, more preferably 5-6 membered ring. Examples of 4 membered heterocycle comprise but not limited to azetidinyl. Examples of 5 membered heterocycle comprise but not  limited to pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidinyl. Examples of 6 membered heterocycle comprise but not limited to piperidinyl, morpholinyl, piperazinyl. Examples of 7 membered heterocycle comprise but not limited to azacycloheptanyl, or the like.
The term “pharmaceutical combination” refers to a combined form of two or more active agents. It will be understood that these agents may be in a mixed or integrated form, e.g. a composition or mixture; or in a separated form, for example in separated compartments of a kit or in different kits. The agents in the pharmaceutical combination may be given in an administration schedule that is synchronous, serial, overlapping, alternating, parallel, or any other treatment schedule in which the various agents are administered as part of a single treatment regimen. The active ingredient is exemplified as one or more agents according to the invention, for example a compound of formula (I) or a pharmaceutically acceptable salt thereof or one or more additional cancer therapeutic agents as mentioned herein.
The term “pharmaceutical composition” refers to an active agent (s) , which is optionally combined with one or more pharmaceutically acceptable components (for example, but not limited to carrier) . The active ingredient is exemplified as one or more agents according to the invention, for example a compound of formula (I) or a pharmaceutically acceptable salt thereof or one or more additional cancer therapeutic agents. In addition to the active ingredients, the pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
The term “pharmaceutically acceptable carrier” refers to those carriers which have no significant irritation and do not impair the bioactivity and property of the active compound. This term may also be understood as inert substance which is administered with active ingredient and is beneficial to the administration thereof. Non-limiting examples include but not limited to any of the following substances which may optionally be approved by Food and Drug Administration for use in human or animal: glidant, sweetening agent, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersant, disintegrant, suspending agent, stabilizing agent, isotonic agent, solvent or emulsifying agent. Other non-limiting examples of the carriers comprise calcium carbonate, calcium phosphate, various sugars and starches, cellulose derivative, gelatine, vegetable oil and polyethylene glycol or the like. Other information regarding the carriers may be found in Remington: The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams &Wilkins (2005) , of which the contents are incorporated herein by reference.
The term “administration” or “administrating” refers to a method that enables a compound, composition or combination to be delivered to a desired site of biological action. Such methods comprise but not limited to oral, parenteral (including intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular injection or infusion) , local, rectal administration or the like.
As used herein, the term “effective amount” refers to the amount of a medicament or agent or combination which is sufficient to achieve the desired effect. The effective amount may be determined individually and depends on the age and general condition of the receptor as well as specific active substance. The effective amount in specific case can be determined by a person skilled in the art through conventional test. When two or more agents are used in combination, for example, in the form of the pharmaceutical combination as claimed, the effective amount also refers to those of each of the agents exerting synergic effect.
The term “active ingredient” , “therapeutic agent” , “active substance” or “active agent” refers to a chemical entity useful for treating or preventing target disorder, disease or condition.
The term “pharmaceutically acceptable” refers to the compound, material, composition, combination and/or dosage form, which are within the scope of reliable medical judgment, suitable for contact with human and animal tissues, without over toxicity, irritation, allergic reaction or other problems or complications and has acceptable benefit/risk ratio.
The term “combined preparation” used herein is defined as especially a "kit of parts" in the sense that the combination partners can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners, i.e., simultaneously or at different time points. The parts of the kit of parts can then, e.g., be administered simultaneously or at different time points and with equal or different time intervals for any part of the kit of parts. 
The terms “cancer” and “cancerous tumor” have the same meaning herein, and include but not limited to solid tumors and blood cancers. Exemplary solid tumors include but not limited to tumors of the breast, glioblastoma, bone, prostate, lung, adrenal gland (e.g., adrenocortical tumors) , bile duct, bladder, bronchus, nervous tissue (including neuronal and glial tumors) , gall bladder, stomach, salivary gland, esophagus, small intestine, cervix, colon, rectum, liver, ovary, pancreas, pituitary adenomas, and secretory adenomas. Exemplary blood cancers include but not limited to lymphomas and leukemia. Exemplary lymphomas include but not limited to multiple myeloma, Hodgkin's lymphoma, non-Hodgkin’s lymphomas (e.g., cutaneous T cell lymphomas such as Sezary syndrome and Mycosis fungoides, diffuse large cell lymphoma, HTLV-1  associated T cell lymphoma, nodal peripheral T cell lymphoma, extranodal peripheral T cell lymphoma, central nervous system lymphoma, and AIDS-related lymphoma) . Exemplary leukemia include but not limited to acute and chronic types of both lymphocytic and myelogenous leukemia (e.g. acute lymphocytic or lymphoblastic leukemia, acute myelogenous leukemia, acute myeloid leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T cell prolymphocyte leukemia, adult T cell leukemia, and hairy cell leukemia) . In a particularly preferable embodiment, the “cancerous tumor” comprises triple negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma or multiple myeloma.
The “beneficial effect” herein for example refers to achieve additional advantageous therapeutic effects, diminish the incidence of side-effects or toxic effects (e.g., diarrhea or nausea) , delay or slow down progression of cancer, reduce the tumor volume in a cancer patient, prolong survival of a cancer patient, prevent or delay tumor metastasis, decrease mortality and morbidity; or to sensitize a cancer patient to the cancer therapeutic agent (s) when an IRE-1α inhibitor is combined with the cancer therapeutic agent (s) ; or to reduce resistance to the cancer therapeutic agent (s) in a cancer patient who has been primarily resistant to such cancer therapeutic agent (s) . In an embodiment, the beneficial effect refers to exerting synergic effect as compared with either of combination partners employed alone.
The term “subject” or “patient” used herein refers to mammal subject or patient, preferably human subject or patient.
Pharmaceutical combination
The present inventor surprisingly found that at least one beneficial effect for treating cancer is observed when the IRE1α inhibitor as a compound of formula (I) or a pharmaceutically acceptable salt thereof is employed in combination therapy, for example one or more additional cancer therapeutic agents as recited herein.
Accordingly, in an aspect of the invention, provided is a pharmaceutical combination comprising (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof and (b) one or more additional cancer therapeutic agents
Figure PCTCN2018096613-appb-000007
wherein
R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1-C 6 hydrocarbon chain containing 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino;
R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6alkoxyl, C 1-6hydroxylalkyl, C 1-6alkoxylC 1-6alkyl,
Figure PCTCN2018096613-appb-000008
and 
Figure PCTCN2018096613-appb-000009
R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6alkoxyl C 1-6alkyl; or
R 9 and R 10 together with the nitrogen atom to which they are attached form 3-10 membered heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6alkylamino, C 1-6 alkoxyl.
In a specific embodiment of the invention, the compound of formula (I) has the following formula (II) :
Figure PCTCN2018096613-appb-000010
It will be understood that the additional cancer therapeutic agent (s) used in the combination according to the invention refers to a therapeutic agent (s) beyond the compound of formula (I) or pharmaceutically acceptable salt thereof as IRE1α inhibitor.
Inositol requiring enzyme-1α (IRE1α) is a transmembrane stress-sensing and signaling molecule that controls the Unfolded Protein Response (UPR) . Numerous perturbations of protein folding contribute to Endoplasmic Reticulum (ER) stress. Downstream enzymatic activity is selectively activated during times of cellular stress, primarily during disease states and thus, inhibition of this pathway may impact tumor growth. Moreover, X-box protein 1 (XBP1) is activated in certain cancer types and may modulate the progression of disease. In vitro data shows that depletion of  XBP1 inhibits tumor growth and relapse. XBP1 splicing activation is up-regulated in cancer and increased following chemotherapy and therefore, is suspected to play a key role in drug resistance. 
The present inventor surprisingly found that various types of physiological stress induce the unfolded protein response including but not limited to hypoxia, nutrient starvation, acidosis, and genetic damage resulting in mutant or over-expressed misfolded proteins (oncogenic stress) and one or more of these conditions are manifest in cancer cells, which may in part be mediated by the microenvironment of the tumor. Without wishing to be bound to a theory, it is believed that the cytoprotective arm of the unfolded protein response (UPR) plays an anti-apoptotic role in tumor survival. In addition, bio-and chemotherapeutic drugs and radiation treatments may further impact the protein folding and degradation cycle in the ER thereby inducing the UPR as a protective resistance mechanism. Patients succumb to cancer because either the tumor is resistant to conventional therapies or returns in a resistant form after an initial response to treatment.
Although the compound of formula (I) or pharmaceutically acceptable salt thereof as IRE1αinhibitor per se can be used as cancer therapeutic agent to some extent, when it is used in combination with other cancer therapeutic agent, the efficacy for treating cancer can be enhanced. Chemotherapeutic agents, targeted small molecule oncology compounds, biomolecule etc. can directly induce ER stress and resulting UPR. IRE1α inhibitors can suppress this activation and thus can act synergistically for cellular proliferation inhibition when used in combination.
Accordingly, in an embodiment of the invention, the additional cancer therapeutic agent (s) has at least one of the following features:
(1) inducing ER stress;
(2) inducing or up-regulating IRE-1α expression;
(3) inducing or up-regulating XBP1 splicing; and
(4) being less effective when IRE-1α is expressed.
In an embodiment according to the invention, the one or more additional cancer therapeutic agents are selected from the group consisting of: cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents, e.g. brefeldin; immunosuppressives; anti-angiogenic compounds and vascular endothelial growth factor (VEGF) inhibitors; fibroblast growth factor (FGF/FGFR) inhibitors; epidermal growth factor receptor (EGFR) inhibitors; antibodies; checkpoint inhibitors;  PD-1 inhibitors or PD-L1 inhibitors; cell cycle inhibitors and differentiation inducers; mTOR inhibitors; corticosteroids; growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers; caspase activators; chromatin disruptors and DNA repair enzyme inhibitors; HDAC inhibitors; Bcr-Abl inhibitors; FMS-like tyrosine kinase 3 (Flt3) inhibitors or any combination thereof.
Some non-limiting examples of the cancer therapeutic agents are as follows:
1) cytotoxic chemotherapeutic agents, including microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) ;
2) antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) ;
3) antimitotic agents such as vinca alkaloids (e.g., eribulin, vinblastine, vincristine, and vinorelbine) ;
4) alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil) , ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa) , alkyl sulfonates-busulfan, nitrosoureas (e.g., carmustine (BCNU) and analogs, streptozocin) , and trazenes-dacarbazinine (DTIC) ;
5) DNA damaging agents such as amsacrine, busulfan, camptothecin, irinotecan (CPT-11) , topotecan, chlorambucil, cyclophosphamide, cytoxan, hexamethylmelamineoxaliplatin, iphosphamide, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, teniposide, triethylenethiophosphoramide and etoposide (VP 16) ;
6) antitumor antibiotics such as actinomycin, dactinomycin (actinomycin D) , daunorubicin, doxorubicin (adriamycin) , epirubicin, idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin;
7) platinum coordination complexes such as cisplatin, carboplatin, and oxaliplatin;
8) proteasome inhibitors, including bortezomib ( [ (1R) -3-methyl-1- [ [ (2S) -1-oxo-3-phenyl-2-[ (pyrazinylcarbonyl) amino] propyl] amino] butyl] boronic acid; MG-341;
Figure PCTCN2018096613-appb-000011
MG-132 (N- [ (phenylmethoxy) carbonyl] -L-leucyl-N- [ (1 S) -1-formyl-3-methylbutyl] -L-leucinamide) , carfilzomib
Figure PCTCN2018096613-appb-000012
and ixazomib
Figure PCTCN2018096613-appb-000013
9) HSP90 inhibitors, including geldanamycin, radicicol, 17AAG, and Gamitrinib;
10) hormones and hormone analogs, including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, bazedoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) ;
11) aromatase inhibitors, e.g. letrozole, anastrozole;
12) fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase) , including aspirin, COX-2 inhibitors, dipyridamole, ticlopidine, clopidogrel, abciximab;
13) antimigratory agents, e.g. somatostatin, wortmannin and PD98059;
14) antisecretory agents, e.g. brefeldin;
15) immunosuppressives, including cyclosporine, tacrolimus (FK-506) , sirolimus (rapamycin) , azathioprine, mycophenolate mofetil;
16) anti-angiogenic compounds (e.g., TNP 470, genistein) and vascular endothelial growth factor (VEGF) inhibitors such as ZD6474, sunitinib, vatalanib, sorafenib, bevacizumab;
17) fibroblast growth factor (FGF/FGFR) inhibitors such as BGJ398, AZD4547, dovitinib, lenvatinib, JNJ-42756493, GP369, BAY1187982;
18) epidermal growth factor receptor (EGFR) inhibitors such as afatinib, gefitinib, erlotinib;
19) antibodies, including trastuzumab
Figure PCTCN2018096613-appb-000014
bevacizumab
Figure PCTCN2018096613-appb-000015
cetuximab 
Figure PCTCN2018096613-appb-000016
rituximab
Figure PCTCN2018096613-appb-000017
20) checkpoint inhibitors, including CTLA4 inhibitors;
21) PD-1 inhibitors or PD-L1 inhibitors such as nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, in particular an antibody or binding fragment thereof;
22) cell cycle inhibitors and differentiation inducers, e.g. tretinoin, ribociclib, palbociclib;
23) mTOR inhibitors, including rapamycin, everolimus, sirolimus, temsirolimus, ridaforolimus; 24) corticosteroids, including cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone;
25) growth factor signal transduction kinase inhibitors such as imatinib, erlotinib, sorafenib, sunitinib, lapatinib, trametinib, temozolomide;
26) mitochondrial dysfunction inducers such as α-tocopherol, Bcl-2 and Bcl-XL inhibitors such as venetoclax, ABT-737, navitoclax, obatoclax mesylate;
27) caspase activators such as 25-hydroxycholesterol, mitomycin C, proscillaridin A, zearalenone, fumonisin B1, garcinol;
28) chromatin disruptors and DNA repair enzyme inhibitors including PARP inhibitors such as 3-aminobenzamide, olaparib, talazoparib, niraparib, veliparib, rucaparib;
29) HDAC inhibitors, e.g. 17-AAG
Figure PCTCN2018096613-appb-000018
suberoylanilide hydroxamic acid
Figure PCTCN2018096613-appb-000019
30) Bcr-Abl inhibitors, including imatinib, nilotinib, dasatinib, bosutinib, ponatinib;
31) FMS-like tyrosine kinase 3 (Flt3) inhibitors, including gilteritinib, lestaurtinib, midostaurin, nintedanib.
In a preferable embodiment, the additional cancer therapeutic agent is selected from the group  consisting of:
Bcr-Abl inhibitors, such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib, particularly nilotinib, dasatinib;
FMS-like tyrosine kinase 3 (Flt3) inhibitors, such as gilteritinib, lestaurtinib, midostaurin, nintedanib, particularly lestaurtinib;
anti-angiogenic compounds and vascular endothelial growth factor (VEGF) inhibitors, such as TNP 470, genistein, ZD6474, sunitinib, vatalanib, sorafenib, bevacizumab, vatalanib; particularly sorafenib,  vatalinib;
epidermal growth factor receptor (EGFR) inhibitors, such as afatinib, gefitinib, erlotinib, particularly gefitinib;
mTOR inhibitors, such as apamycin, everolimus, sirolimus, temsirolimus, ridaforolimus, particularly  temsirolimus;
HDAC inhibitors, such as 17-AAG
Figure PCTCN2018096613-appb-000020
vorinostat
Figure PCTCN2018096613-appb-000021
particularly  vorinostat
cytotoxic chemotherapeutic agents, such as microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) , particularly taxane (e.g. paclitaxel, docetaxel) ;
antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) , particularly gemcitabine;
proteasome inhibitors, such as bortezomib, MG-132, carfilzomib, ixazomib, particularly bortezomib;
hormones and hormone analogs, including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, bazedoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) , particularly tamoxifen.
In a preferable embodiment, the additional cancer therapeutic agent is selected from the group consisting of cytotoxic chemotherapeutic agent, proteasome inhibitor, hormone analogue or any combination thereof. More preferably, the cytotoxic chemotherapeutic agent is selected from the group consisting of microtubule disruptors, e.g. paclitaxel or docetaxel. More preferably, the proteasome inhibitor is bortezomib. More preferably, the hormone analogue is anti-estrogen agent, e.g. tamoxifen.
In a preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and one cytotoxic chemotherapeutic  agent.
In a more preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor.
In an even more preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor selected from paclitaxel or docetaxel.
In a most preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and paclitaxel.
In a preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and a proteasome inhibitor.
In a more preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and bortezomib.
In a preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and a hormone analogue.
In a more preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and an estrogen receptor inhibitor.
In an even more preferable embodiment, provided is a pharmaceutical combination, comprising a compound of formula (II) or a pharmaceutically acceptable salt thereof and tamoxifen.
It will be understood that the agents in the pharmaceutical combination according to the invention, either the compound of formula (I) or the additional cancer therapeutic agent (s) , encompass their other forms like stereoisomers, salts, prodrugs as well as crystal modifications, e.g. solvates and polymorphs and such forms are within the scope of the present invention. Preferably, these forms are pharmaceutically acceptable.
The effective amounts or dosages of the compound of formula (I) or the pharmaceutically  acceptable salt thereof and one or more additional cancer therapeutic agents employed in the pharmaceutical combination according to the invention may vary depending on the particular compound or agent (s) employed, the mode of administration, the condition being treated, and severity of the condition being treated etc. Thus, the dosage regimen is selected in accordance with a variety of factors including the route of administration, the renal and hepatic function of the subject or the like. A physician, clinician or veterinarian of ordinary skill can readily determine and prescribe the effective amount required to prevent, counter or arrest the progress of the condition. Typically, the effective dosage of the compound of formula (I) or the pharmaceutically acceptable salt thereof for daily use is about 10-2000 mg, preferably 50-1000 mg for a warm-blooded animal like human of about 70 kg bodyweight. The effective dosage of the one or more additional cancer therapeutic agents for daily use in a warm-blooded animal, including man, can be determined by a package insert when said agent is provided as a marketed drug. It might be also be possible that the effective dosage of the one or more additional cancer therapeutic agents is adjusted according to species, age, individual condition, mode of administration, the clinical picture in question, etc.
In a particular embodiment, the pharmaceutical combination of the invention comprises
1) the compound of formula (II) or the pharmaceutically acceptable salt thereof
Figure PCTCN2018096613-appb-000022
and
2) one or more additional cancer therapeutic agents.
In an embodiment of the invention, the additional cancer therapeutic agents are defined as above.
Pharmaceutical composition and kits
The pharmaceutical combination according to the invention can further comprise one or more pharmaceutically acceptable carriers. In an embodiment wherein the pharmaceutical combination is provided in a unique form such as a pharmaceutical composition or mixture, the compound or agent (s) contained therein are combined with the same pharmaceutically acceptable carriers, for simultaneous, separate or sequential use.
The pharmaceutical composition according to the invention can be prepared in a manner known per se and are those suitable for enteral, such as oral or rectal, and parenteral administration to  mammals (warm-blooded animals) , including man, comprising a therapeutically effective amount of compound of formula (I) and at least a therapeutically effective amount of cancer therapeutic agent, or further in combination with one or more pharmaceutically acceptable carries, especially suitable for enteral or parenteral application.
In an alternative embodiment wherein the pharmaceutical combination is provided in separated forms such as different compartments in a kit or in different kits, the agents contained therein, either the compound of formula (I) or the additional cancer therapeutic agent (s) , are independently combined with the pharmaceutically acceptable carriers. The pharmaceutically acceptable carriers for each of the agent may be identical or different according to practice requirement. Accordingly, provided is also a kit, comprising (a) a compound of formula (I) or pharmaceutically acceptable salt thereof and optional one or more pharmaceutically acceptable carriers; (b) one or more additional cancer therapeutic agents and optional one or more pharmaceutically acceptable carriers; and (c) instruction for using (a) and (b) . The compound of formula (I) or pharmaceutically acceptable salt thereof and the additional cancer therapeutic agents are defined as above.
The ratio of the total amounts of the compound of formula (I) or the pharmaceutically acceptable salt thereof to one or more additional cancer therapeutic agents in the pharmaceutical combination according to the invention can be varied, e.g. in order to cope with the needs of a patient sub-population to be treated or the needs of a single patient which different needs can be due to the particular disease, age, sex, body weight, etc.
Methods and use according to the invention
In another aspect according to the invention, provided is a method for treating cancerous tumor, comprising administering a subject in need thereof an effective amount of the pharmaceutical combination according to the invention, wherein the active agents comprised in the pharmaceutical combination are defined as above.
In an embodiment of the method of the invention, the pharmaceutical combination comprises (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof and (b) one or more additional cancer therapeutic agents:
Figure PCTCN2018096613-appb-000023
wherein R 3, R 4, R 5 and R 6 are defined as above.
In a specific aspect of the invention, provided is a method for treating cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents:
Figure PCTCN2018096613-appb-000024
wherein R 3, R 4, R 5 and R 6 are defined as above.
In an embodiment of the methods above, the one or more additional cancer therapeutic agents are defined as above. In a specific embodiment, the cancer therapeutic agent has at least one of the following features: (1) inducing ER stress; (2) inducing or up-regulating IRE-1α expression; (3) inducing or up-regulating XBP1 splicing; and (4) being less effective when IRE-1α is expressed.
In some embodiments, said treatment is to cure the disease or to have an effect on disease regression or on the delay of progression of the disease. In an embodiment, said treatment is to inhibit the growth of tumor, for example, to reduce tumor volume, to delay the growth of tumor, to reverse the growth of tumor or any combination thereof. In another embodiment, said treatment is to kill the tumor, for example, to maintain the growth under a very low level.
Upon administration of the pharmaceutical combination according to the invention, the agents comprised therein (either compound of formula (I) or pharmaceutically acceptable salt thereof or one or more additional cancer therapeutic agents) are intended for simultaneous, separate or sequential use. Alternatively, upon administration of compound of formula (I) or pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents, such agents are intended for simultaneous, separate or sequential use.
For example, the compound of formula (I) or pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents can be used, e.g. as a combined preparation or a pharmaceutical composition/mixture such that they can be administered at essentially the same time. Alternatively, the compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents can be in different compartments of a kit or  different kits such that they can be administered at different time. For administration at different time, mention can be given to an order according to practical requirement. The compound of formula (I) or the pharmaceutically acceptable salt thereof can be administered before, after or along with the additional cancer therapeutic agent. The time interval between administrations of these agents may be several minutes, hours, days, months or even longer according to practical requirement.
Moreover, when the agents in the combination according to the invention are administered at different time points, the time intervals are such that the effect on the treated cancer in the combined use is larger than the effect which would be obtained by use of only any one of the combination partners.
If needed, the agents in the pharmaceutical combination according to the invention may be administered in the same or different routes. For example, the compound of formula (I) or pharmaceutically acceptable salt thereof and the additional cancer therapeutic agent (s) may be both administered orally or intravenously. Alternatively, the compound of formula (I) or pharmaceutically acceptable salt thereof may be administered orally while the additional cancer therapeutic agent (s) may be administered intravenously and vice versa. It would be understood that when more than one additional cancer therapeutic agents are used, their administration routes are selected independently, i.e. either identical or different.
In a specific embodiment according to the methods above, the compound of formula (I) has the following formula (II) :
Figure PCTCN2018096613-appb-000025
In yet a further aspect of the invention, provided is a method for enhancing the efficacy of a cancer therapeutic agent, comprising applying a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with the cancer therapeutic agent;
Figure PCTCN2018096613-appb-000026
wherein R 3, R 4, R 5 and R 6 are defined as above.
In a specific embodiment of the invention, the compound of formula (I) has the following formula (II) :
Figure PCTCN2018096613-appb-000027
In an embodiment of the invention, the cancer therapeutic agent has at least one of the following features:
(1) inducing ER stress;
(2) inducing or up-regulating IRE-1α expression;
(3) inducing or up-regulating XBP1 splicing; and
(4) being less effective when IRE-1α is expressed.
In an embodiment of the invention, the enhancement of efficacy is embodied in inhibiting the growth of tumor, for example, reducing tumor volume, delaying the growth of tumor, reversing the growth of tumor or any combination thereof. Alternatively, enhancement of efficacy is embodied in killing the tumor, for example, maintaining the growth under a very low level.
In an embodiment of the invention, the one or more additional cancer therapeutic agents or the cancer therapeutic agent of which the efficacy to be enhanced are selected from the group consisting of cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents, e.g. brefeldin; immunosuppressives; anti-angiogenic compounds and vascular endothelial growth factor (VEGF) inhibitors; fibroblast growth factor (FGF/FGFR) inhibitors; epidermal growth factor receptor (EGFR) inhibitors; antibodies; checkpoint inhibitors; PD-1 inhibitors or PD-L1 inhibitors; cell cycle inhibitors and differentiation inducers; mTOR inhibitors; corticosteroids; growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers; caspase activators; chromatin disruptors and DNA repair enzyme inhibitors; HDAC inhibitors; Bcr-Abl inhibitors; FMS-like tyrosine kinase 3 (Flt3) inhibitors or any combination thereof.
Some examples of the cancer therapeutic agents are as follows:
1) cytotoxic chemotherapeutic agents, including microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) ;
2) antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) ;
3) antimitotic agents such as vinca alkaloids (e.g., eribulin, vinblastine, vincristine, and vinorelbine) ;
4) alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil) , ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa) , alkyl sulfonates-busulfan, nitrosoureas (e.g., carmustine (BCNU) and analogs, streptozocin) , and trazenes-dacarbazinine (DTIC) ;
5) DNA damaging agents such as amsacrine, busulfan, camptothecin, irinotecan (CPT-11) , topotecan, chlorambucil, cyclophosphamide, cytoxan, hexamethylmelamineoxaliplatin, iphosphamide, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, teniposide, triethylenethiophosphoramide and etoposide (VP 16) ;
6) antitumor antibiotics such as actinomycin, dactinomycin (actinomycin D) , daunorubicin, doxorubicin (adriamycin) , epirubicin, idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin;
7) platinum coordination complexes such as cisplatin, carboplatin, and oxaliplatin;
8) proteasome inhibitors, including bortezomib ( [ (1R) -3-methyl-1- [ [ (2S) -1-oxo-3-phenyl-2- [ (pyrazinylcarbonyl) amino] propyl] amino] butyl] boronic acid; MG-341;
Figure PCTCN2018096613-appb-000028
) , MG-132 (N- [ (phenylmethoxy) carbonyl] -L-leucyl-N- [ (1 S) -1-formyl-3-methylbutyl] -L-leucinamide) , carfilzomib
Figure PCTCN2018096613-appb-000029
and ixazomib
Figure PCTCN2018096613-appb-000030
9) HSP90 inhibitors, including geldanamycin, radicicol, 17AAG, and gamitrinib;
10) hormones and hormone analogs, including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, bazedoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) ;
11) aromatase inhibitors, e.g. letrozole, anastrozole;
12) fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase) , including aspirin, COX-2 inhibitors, dipyridamole, ticlopidine, clopidogrel, abciximab;
13) antimigratory agents, e.g. somatostatin, wortmannin and PD98059;
14) antisecretory agents, e.g. brefeldin;
15) immunosuppressives, including cyclosporine, tacrolimus (FK-506) , sirolimus (rapamycin) , azathioprine, mycophenolate mofetil;
16) anti-angiogenic compounds (e.g., TNP 470, genistein) and vascular endothelial growth factor (VEGF) inhibitors such as ZD6474, sunitinib, vatalanib, sorafenib, bevacizumab;
17) fibroblast growth factor receptor (FGF/FGFR) inhibitors such as BGJ398, AZD4547, dovitinib, lenvatinib, JNJ-42756493, GP369, BAY1187982;
18) epidermal growth factor (EGFR) inhibitors such as afatinib, gefitinib, erlotinib;
19) antibodies, including trastuzumab
Figure PCTCN2018096613-appb-000031
bevacizumab
Figure PCTCN2018096613-appb-000032
cetuximab 
Figure PCTCN2018096613-appb-000033
rituximab
Figure PCTCN2018096613-appb-000034
20) checkpoint inhibitors, including CTLA4 inhibitors,
21) PD-1 inhibitors or PD-L1 inhibitors such as nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, in particular an antibody or binding fragment thereof;
22) cell cycle inhibitors and differentiation inducers, e.g. tretinoin, ribociclib, palbociclib;
23) mTOR inhibitors, including rapamycin, everolimus, sirolimus, temsirolimus, ridaforolimus;
24) corticosteroids, including cortisone, dexamethasone, hydrocortisone, methylpednisolone,prednisone, and prenisolone;
25) growth factor signal transduction kinase inhibitors such as imatinib, erlotinib, sorafenib, sunitinib, lapatinib, trametinib, temozolomide;
26) mitochondrial dysfunction inducers such as α-tocopherol, Bcl-2 and Bcl-XL inhibitors such as venetoclax, ABT-737, navitoclax, obatoclax mesylate;
27) caspase activators such as 25-hydroxycholesterol, mitomycin C, proscillaridin A, zearalenone, fumonisin B1, garcinol;
28) chromatin disruptors and DNA repair enzyme inhibitors including PARP inhibitors such as 3-aminobenzamide, olaparib, talazoparib, niraparib, veliparib, rucaparib;
29) HDAC inhibitors, e.g. 17-AAG
Figure PCTCN2018096613-appb-000035
suberoylanilide hydroxamic acid
Figure PCTCN2018096613-appb-000036
30) Bcr-Abl inhibitors, including imatinib, nilotinib, dasatinib, bosutinib, ponatinib;
31) FMS-like tyrosine kinase 3 (Flt3) inhibitors, including gilteritinib, lestaurtinib, midostaurin, nintedanib.
In a preferable embodiment of the invention, the additional cancer therapeutic agent is selected from the group consisting of:
Bcr-Abl inhibitors, such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib, particularly nilotinib, dasatinib;
FMS-like tyrosine kinase 3 (Flt3) inhibitors, such as gilteritinib, lestaurtinib, midostaurin, nintedanib, particularly lestaurtinib;
anti-angiogenic compounds and vascular endothelial growth factor (VEGF) inhibitors, such as TNP 470, genistein, ZD6474, sunitinib, vatalanib, sorafenib, bevacizumab, vatalanib; particularly  sorafenib,  vatalinib;
epidermal growth factor receptor (EGFR) inhibitors, such as afatinib, gefitinib, erlotinib, particularly gefitinib;
mTOR inhibitors, such as apamycin, everolimus, sirolimus, temsirolimus, ridaforolimus, particularly  temsirolimus;
HDAC inhibitors, such as 17-AAG
Figure PCTCN2018096613-appb-000037
vorinostat
Figure PCTCN2018096613-appb-000038
particularly  vorinostat
cytotoxic chemotherapeutic agents, such as microtubule disruptors such as taxane (paclitaxel, docetaxel) , vincristin, vinblastin, nocodazole, epothilones and navelbine, and epipodophyllotoxins (e.g., teniposide) , particularly taxane (paclitaxel, docetaxel) ;
antimetabolites such as pyrimidine analogs (e.g., 5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) , purine analogs, folate antagonists and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine) , and folic acid analogs (e.g., methotrexate) , particularly gemcitabine;
proteasome inhibitors, such as bortezomib, MG-132, carfilzomib, ixazomib, particularly bortezomib;
hormones and hormone analogs, including estrogen, goserelin, estrogen receptor inhibitors (e.g. raloxifene, tamoxifen, bazedoxifene) , androgen receptor inhibitors (e.g. bicalutamide, nilutamide, enzalutamide) , particularly tamoxifen.
In a preferable embodiment of the invention, the additional cancer therapeutic agent used in the method for treatment of cancerous tumor or the cancer therapeutic agent of which the efficacy to be enhanced is selected from the group consisting of cytotoxic chemotherapeutic agent, proteasome inhibitor, hormone analogue or any combination thereof. More preferably, the cytotoxic chemotherapeutic agent is selected from the group consisting of microtubule disruptors, e.g. paclitaxel or docetaxel. More preferably, the proteasome inhibitor is bortezomib. More preferably, the hormone analogue is anti-estrogen agent, e.g. tamoxifen.
In a preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and one cytotoxic chemotherapeutic agent.
In a more preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor.
In an even more preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and one microtubule disruptor selected from paclitaxel or docetaxel.
In a most preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and paclitaxel.
In a preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and a proteasome inhibitor.
In a more preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and bortezomib.
In a preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and a hormone analogue.
In a more preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and an estrogen receptor inhibitor.
In an even more preferable embodiment, provided is a method for treatment of cancerous tumor, comprising administering a subject in need thereof an effective amount of a compound of formula (II) or a pharmaceutically acceptable salt thereof and tamoxifen.
In yet a further aspect, provided is the pharmaceutical combination according to the invention for treating cancerous tumor.
In still a further aspect, provided is use of the pharmaceutical combination according to the invention for the manufacture of a medicament for treating cancerous tumor.
In an embodiment, the pharmaceutical combination comprises (a) a compound of formula (I) or a  pharmaceutically acceptable salt thereof and (b) one or more additional cancer therapeutic agents:
Figure PCTCN2018096613-appb-000039
wherein R 3, R 4, R 5 and R 6 are defined as above.
Preferably, the compound of formula (I) has the following formula (II) :
Figure PCTCN2018096613-appb-000040
Preferably, the additional cancer therapeutic agents are defined as above.
Cancerous tumor
The cancerous tumors which can be treated with the pharmaceutical combination according to the invention or to which the efficacy of the cancer therapeutic agent can be enhanced comprise but not limited to solid tumors and blood cancers.
Exemplary solid tumors include, but are not limited to tumors of breast, glioblastoma, bone, prostate, lung, adrenal gland (e.g., adrenocortical tumors) , bile duct, bladder, bronchus, nervous tissue (including neuronal and glial tumors) , gall bladder, stomach, salivary gland, esophagus, small intestine, cervix, colon, rectum, liver, ovary, pancreas, pituitary adenomas, and secretory adenomas. Blood cancers include lymphomas and leukemia. Exemplary lymphomas include, but are not limited to multiple myeloma, Hodgkin's lymphoma, non-Hodgkin’s lymphomas (e.g., cutaneous T cell lymphomas such as Sezary syndrome and Mycosis fungoides, diffuse large cell lymphoma, HTLV-1 associated T cell lymphoma, nodal peripheral T cell lymphoma, extranodal peripheral T cell lymphoma, central nervous system lymphoma, and AIDS-related lymphoma) . Exemplary leukemia include, but are not limited to acute and chronic types of both lymphocytic and myelogenous leukemia (e.g. acute lymphocytic or lymphoblastic leukemia, acute myelogenous leukemia, acute myeloid leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T cell prolymphocyte leukemia, adult T cell leukemia, and hairy cell leukemia) .
In some embodiments, the cancer is selected from  liver cancer, breast cancer, lung cancer, ovarian cancer, head and neck cancer, glioblastoma, and multiple myeloma.
In a preferable embodiment, the breast cancer is triple negative breast cancer. More preferably, triple negative breast cancer is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, the breast cancer is estrogen positive breast cancer. More preferably, estrogen positive breast cancer is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, the lung cancer is non-small cell lung carcinoma (NSCLC) . More preferably, non-small cell lung carcinoma is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, the lung cancer is small cell lung carcinoma (SCLC) . More preferably, small cell lung carcinoma is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, the ovarian cancer is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, the head and neck cancer is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, glioblastoma is metastatic, recurrent, refractory or advanced.
In a preferable embodiment, multiple myeloma is metastatic, recurrent, refractory or advanced.
Beneficial effect
The present inventor has performed a 7-day toxicity study based on cynomolgus monkey model showed that Orin1001 as the compound of formula (I) or pharmaceutically acceptable salt thereof has a No Observable Aderse Effect Level (NOAEL) of 150 mg/kg/d, and a 5-fold safety margin amounting to 750 mg/kg/d, which indicates a good safety profile of such compound and thus it can be used in a relatively high dosage without significant side effect like toxicity, either alone or in combination with other cancer therapeutical agent. Accordingly, the pharmaceutical combination according to the invention can be used to effectively treat cancer/tumor by inhibiting tumor growth or killing tumor, for example delaying, arresting, or reversing tumor growth with synergist effect and good safety.
Examples
Some Abbreviations: PO: oral; sc: subcutaneous; iv: intravenous; qod: every other day; qwk: once a week; qd: once a day.
In addition to UPR activation leading to XBP1 splicing by a myriad of cellular insults including genotoxic and microenvironmental stresses, IRE1 can be activated indirectly by a number of small molecules. In the Examples, we explored a number of mechanistically distinct FDA approved or clinical level oncology compounds and surprisingly found that many can induce XPB1 splicing.
The materials and reagents used in the Examples are commercially available. The cells are available from ATCC (American type culture collection) . MM. 1S (
Figure PCTCN2018096613-appb-000041
CRL-2974 TM) is human plasmacytoma/myeloma cell; HEK-293 (
Figure PCTCN2018096613-appb-000042
CRL-1573 TM) is human embryonic kidney cell; H929 (
Figure PCTCN2018096613-appb-000043
CRL-9068 TM) is human plasmacytoma/myeloma; RPMI8226 (
Figure PCTCN2018096613-appb-000044
CCL-155 TM) is human plasmacytoma/myeloma cell; A549 (
Figure PCTCN2018096613-appb-000045
CCL-185 TM) is human Lung epithelial carcinoma cell; HT-29 (
Figure PCTCN2018096613-appb-000046
HTB-38 TM) is human colorectal adenocarcinoma; MCF7 (
Figure PCTCN2018096613-appb-000047
HTB-22 TM) is human epithelial mammary gland adenocarcinoma; and Hep G2 (
Figure PCTCN2018096613-appb-000048
HB-8065 TM) is “Hepatoma” , human hepatocellular carcinoma. Unless stated otherwise, the apparatus and reagents are available from Invitrogen.
Examples 1-12 were carried out by the method described below.
Determination of XBP1s level induced by various compounds:
This method applies to any mammalian cell line but was typically applied to human MM1s myeloma cells for EC 50 and RPMI8826 plasmacytoma cells for confirmation of selected compounds. Briefly, cells were grown in standard conditions and spread into 96 well tissue culture plates. Cells were treated with compounds with indicated concentration using serial dilutions. DTT (dithiothreitol) or compounds were added at the same time and cells were harvested after indicated hour’s treatment. Cells treated with DTT alone were used as 100% XBP1s positive controls and cell left untreated were used as base line XBP1s level.
Splicing Assay: Total RNA was isolated from cells treated with compounds using Applied Biosystems RNAqueous kit. 1 μg of total RNA was reverse transcribed using Oligo dt (12-18) (Invitrogen) . The cDNA was then amplified at 95℃ for 8 min and 30 sec, then 40 cycles at 95℃ for 15 sec and 63℃ for 1 min. Samples were run against a purified standard curve for both spliced and unspliced XBP1, and further normalized to internal house-keeping gene GAPDH.
Probes and primers:
Human XBP1 Forward-GGAAGCCAAGGGGAATGAAGTG (Seq ID No. 1) 
Human XBP1 Reverse-GGAGATGTTCTGGAGGGGTGAC (Seq ID No. 2) 
GAPDH Forward-ATCGTGGAAGGACTCATGACCA (Seq ID No. 3) 
GAPDH Reverse-AGGGATGATGTTCTGGAGAGCC (Seq ID No. 4) 
Human Unspliced XBP1
Probe -5’ CAL FLURO RED-CACGTAG TCTGAGTGCTGCGGACT-BHQ2 3’ (Seq ID No. 5) 
Human Spliced XBP1 Probe-5’ FAM-CCTGCACCTGCTGCGGACT-BHQ1 3’ (Seq ID No. 6) 
GAPDH Probe-5’ HEX-TCCATGCCATCACTGCCACCCA-BHQ1 3’ (Seq ID No. 7) 
Example 1-12
In Examples 1-12, various compounds were tested for their respective induction to cells lines’ ER stress measured by XBP1s level and the results are shown in Figures 1-12, respectively. We have tested the compounds as several FDA approved kinase inhibitors could induce XBP1s: Nilotinib, Sorafinib, Gefitinib, Dasatinib and the late clinical stage kinase inhibitors Vatalinib and Lestaurtinib; as well as the compounds Temsirolimus 
Figure PCTCN2018096613-appb-000049
a FDA approved natural product mTOR inhibitor; Vorinostat, a FDA approved HDAC inhibitor; Paclitaxel, a well characterized microtubule stabilizer; Gemcitibine, a nucleoside analogue; and 17-AAG, a HSP90 inhibitor and shown that they were all able to induce XBP1s in a time, cell or concentration dependent manner.
Example 1 showed that Lestaurtinib enhanced multiple cell lines’ ER stress measured by XBP1s level (Figure 1) .
Example 2 showed that Nilotinib enhanced MM1c cell’s ER stress measured by XBP1s level (Figure 2) . The same methods were used as above for Lestaurtinib.
Example 3 showed that Sorafenib enhanced A549 cell lines’ ER stress measured by XBP1s level (Figure 3) . The same methods were used as above for Lestaurtinib.
Example 4 showed that Dasatinib enhanced A549 cell lines’ ER stress measured by XBP1s level (Figure 4) . The same methods were used as above for Lestaurtinib.
Example 5 showed that Gefitinib enhanced A549 cell lines’ ER stress measured by XBP1s level (Figure 5) . The same methods were used as above for Lestaurtinib.
Example 6 showed that Lestaurtinib, temisirolimus, vatalinib enhanced several cell lines’ ER stress measured by XBP1s level (Figure 6) . The same methods were used as above for Lestaurtinib.
Example 7 showed that Torisel
Figure PCTCN2018096613-appb-000050
 (Temsirolimus) enhanced A549 cell lines’ ER stress measured by XBP1s level (Figure 7) . The same methods were used as above for Lestaurtinib.
Example 8 showed that Vorinostat enhanced HT-29 cell lines’ ER stress measured by XBP1s level (Figure 8) . The same methods were used as above for Lestaurtinib.
Example 9 showed that Paclitaxel enhanced RPMI 8226 cell lines’ ER stress measured by XBP1s level (Figure 9) . The same methods were used as above for Lestaurtinib.
Example 10 showed that Gemcitabine enhanced RPMI 8226 cell lines’ ER stress measured by XBP1s level (Figure 10) . The same methods were used as above for Lestaurtinib.
Example 11 showed that 17-AAG enhanced MCF-7 cell lines’ ER stress measured by XBP1s level (Figure 11) . The same methods were used as above for Lestaurtinib.
Example 12 showed that 17-AAG enhanced Hepatoma cell lines’ ER stress measured by XBP1s level (Figure 12) . The same methods were used as above for Lestaurtinib.
Example 13 showed Intratumoral XBP-1 spliced effect of IRE-1 compound Orin1001 in 
Figure PCTCN2018096613-appb-000051
treated RPMI xenografts (Figure 13) . As shown in Figure 13, Nude mice with xenografts using RPMI 8226 tumor cells were treated by IV injection of 
Figure PCTCN2018096613-appb-000052
at 0.8 mg/kg after the tumor established in 21 days. The mice were treated again on the 24 th day with 
Figure PCTCN2018096613-appb-000053
On Day 27, the mice were treated with Orin1001 at 30 mg/kg PO, four hours later the mice were sacrificed as in Example 22, and tumor tissues were isolated. In a similar way as described for the liver PD test in Example 22, RNA extraction and RT-PCR analysis gave the results as shown Figure 13. This experiment clearly demonstrated that
Figure PCTCN2018096613-appb-000054
increased the level of XBP1s (lower band in the gel image) in tumor which is an indication of IRE1 activation, and Orin1001 can inhibit the activity of the activated IRE1. The results suggest a combined strategy of treating cancer patient with
Figure PCTCN2018096613-appb-000055
and Orin1001.
Examples 14-22
Examples 14-22 were in vivo tests for efficacies of ORIN 1001 in combination with other cancer therapeutic agents and the procedures were summarized as follows.
Administration of an IRE-1α inhibitor in combination with a cytotoxic agent or hormone antagonist may be more effective in inhibiting tumor growth and prevent tumor relapse. A novel, first-in-class IRE-1α inhibitor, Orin1001, was evaluated in combination with paclitaxel, tamoxifen or
Figure PCTCN2018096613-appb-000056
in mouse tumor xenograft models, which were developed by Charles River Laboratories and the studies were performed by Charles River Laboratories as contracted services. These models include triple negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma and multiple myeloma. All the therapeutic agents such as paclitaxel, tamoxifen and 
Figure PCTCN2018096613-appb-000057
were purchased by Charles River from commercial sources.
Example 14: Orin1001 inhibits triple negative breast cancer in combination with paclitaxel at  different tumor growth stage
Orin1001 was administered by oral gavage in combination with paclitaxel in a xenograft mouse model using female NCr nu/nu mice injected subcutaneously with human breast adenocarcinoma MDA-MB231 tumor cells. To evaluate the effect of Orin1001 in combination with paclitaxel on early, mid or late stage tumor growth, dosing was initiated either on Day 1 (when tumors reached 225-250 mm 3) , Day 14 or Day 28 of tumor growth. Orin1001 was administered via oral gavage at 300 mg/kg/day in combination with paclitaxel at 10 mg/kg iv, weekly (n=10/group) for up to 60 days. There were no clinical signs of toxicity with Orin1001. The results are shown in Figure 14.
In combination with paclitaxel, Orin1001 showed significant tumor inhibition compared to paclitaxel alone at all stages of tumor growth. Specifically, Orin1001 (300mg/kg) was applied at Day 1, Day 14 and Day 28 respectively in combination with paclitaxel (10 mg/kg) showing the effects of intervene of Orin1001 at any stage of tumor growth.
As compared to control and paclitaxel alone, the combined use of Orin1001 and paclitaxel could delay the growth of tumor and the synergist effect could be seen at every intervening stage like early, middle or late stage, even starting as late as Day 28. Moreover, also compared to control and paclitaxel alone, the growth of tumor could be reversed when Orin1001 is applied together with paclitaxel at every intervening stage, even starting as late as Day 28. And extended oral dosing of Orin1001 for up to 60 consecutive days was well tolerated and also resulted in a significant synergist effect on tumor inhibition.
Example 15: Orin1001 inhibits triple negative breast cancer in combination with Paclitaxel dose  proportionally
Orin1001 was administered by oral gavage in combination with paclitaxel in a xenograft mouse model using female NCr nu/nu mice (n=10 in each group) injected subcutaneously with human breast adenocarcinoma MDA-MB231 tumor cells. To evaluate the effect of Orin1001 in combination with paclitaxel, Orin1001 was administered via oral gavage at 75, 150 or 300 mg/kg/day in combination with paclitaxel at 10 mg/kg iv, weekly (n=10/group) for up to 60 days. There were no clinical signs of toxicity with Orin1001. The results are shown in Figure 15.
In combination with paclitaxel, Orin1001 showed significant tumor inhibition compared to paclitaxel alone at all stages of tumor growth. Treatment with 300 mg/kg Orin1001 in combination with paclitaxel resulted in 3 partial regressions and 1 tumor-free survival versus 1 partial regression in the paclitaxel group alone. Particularly, when Orin1001 was applied at a dose at 150 mg/kg/day or more, the inhibitory effects were much obvious. At every dosing level of combined use of Orin1001, the growth of tumors could be reversed. Especially, when Orin1001 was applied at a dose of 300 mg/kg, the tumor growth was almost arrested. And extended oral dosing of Orin1001 for up to 60 consecutive days was well tolerated and resulted in a significant synergist effect on tumor inhibition.
Example 16: Orin1001 inhibits estrogen positive breast cancer in combination with tamoxifen Orin1001 was administered by oral gavage alone and in combination with tamoxifen using female NCr nu/nu mice injected with human breast adenocarcinoma MCF-7 tumor cells in an orthotopic mouse xenograft model. Three days prior to tumor cell implantation, estrogen pellets were implanted subcutaneously. Tumor cells used for implantation were harvested during log phase growth and implanted into the mammary fat pad. Tumor growth was monitored as the average size approached the target range of 225-250 mm 3. Orin1001 was administered via oral gavage at 300 mg/kg/day and in combination with tamoxifen at 30μg mg/kg sc, every other day (n=12/group) . The results are shown in Figure 16.
Orin1001 in combination with tamoxifen showed significant tumor inhibition compared to tamoxifen alone.
Example 17: Orin1001 inhibits estrogen positive breast cancer in combination with paclitaxel Orin1001 was administered by oral gavage alone and in combination with paclitaxel using female NCr nu/nu mice injected with human breast adenocarcinoma MCF-7 tumor cells in an orthotopic mouse xenograft model. Three days prior to tumor cell implantation, estrogen pellets were  implanted subcutaneously. Tumor cells used for implantation were harvested during log phase growth and implanted into the mammary fat pad. Tumor growth was monitored as the average size approached the target range of 225–250 mm 3. Orin1001 was administered via oral gavage at 300 mg/kg/day and in combination with paclitaxel at 10 mg/kg iv weekly (n=11/group) . The results are shown in Figure 17.
Orin1001 in combination with paclitaxel showed significant synergistic effects on tumor growth inhibition compared to paclitaxel alone. Specifically, when Orin1001 was applied in combination with paclitaxel, a synergistic effect could be seen over Orin1001 or paclitaxel alone and particularly the growth of tumor was almost arrested and then reversed under the combined use.
Example 18: Orin1001 inhibits ovarian cancer in combination with paclitaxel
Orin1001 was administered by oral gavage in a xenograft mouse model using female NCr nu/nu mice injected subcutaneously with human ovarian carcinoma A2780 tumor cells. Animals were assigned to 4 groups (n=6/group) ; Vehicle control administered by oral gavage for 28 days, Orin1001 administered via oral gavage at 300 mg/kg/day for 28 consecutive days, paclitaxel administered weekly by iv at 15 mg/kg, or Orin1001 administered in combination with paclitaxel. The study endpoint was tumor volume of 2000 mm 3 or Day 60, whichever came first and the results are shown in Figure 18.
The percent tumor growth delay was calculated using the following equation: TGD (%) = [T-C/C] x 100, where T-C is the difference in time to tumor endpoint from Treated (T) and Control (C). The percent TGD was 29, 40 and 68% for Orin1001 alone, paclitaxel alone and Orin1001 in combination with paclitaxel, respectively. According to Figure 18, it can be seen that in combination with Paclitaxel, 300 mg/kg Orin1001 showed increased tumor inhibition compared to paclitaxel alone and the reverse of tumor growth was observed under the combined use of Orin1001 and paclitaxel.
Example 19: Orin1001 inhibits glioblastoma in combination with paclitaxel
Orin1001 was administered by oral gavage in a xenograft mouse model using female NCr nu/nu mice injected subcutaneously with human glioblastoma U-87 MG tumor cells. Animals were assigned to 4 groups (n=6/group) ; Vehicle control administered by oral gavage for 28 days, Orin1001 administered via oral gavage at 300 mg/kg/day for 28 consecutive days, paclitaxel administered weekly by iv at 15 mg/kg, or Orin1001 administered in combination with paclitaxel. The study endpoint was tumor volume of 2000 mm 3 or Day 60, whichever came first and the results are shown in Figure 19.
The percent tumor growth delay was calculated using the following equation: TGD (%) = [T-C/C] x100; where T-C is the difference in time to tumor endpoint from Treated (T) and Control (C). The percent TGD was 13, 17 and 50% for Orin1001 alone, paclitaxel alone and Orin1001 in combination with paclitaxel, respectively.
The time to tumor endpoint (TTE) for each animal was further calculated using the following equation: TTE (days) = log10 (endpoint volume, mm 3) -b /m, where b is the intercept and m is the slope of the line obtained by linear regression of the log-transformed tumor growth data set. The TTE was 25.9, 29.3, 30.2 and 38.0 for vehicle control, Orin1001 alone, paclitaxel alone, Orin1001 in combination with paclitaxel, respectively.
Tumor growth delay and survival were significantly greater with Orin1001 in combination with paclitaxel than with paclitaxel alone (p<0.01, Chi Square and Gehan-Breslow-Wilcoxon test) . In combination with paclitaxel, 300 mg/kg Orin1001 showed a marked increase in tumor inhibition compared to paclitaxel alone with 2 animals showing partial regression versus 0 animals in the other treated groups. Specifically, the tumor delay effects was not very significant for Orin1001 or paclitaxel each alone over the control and the tumor volumes reach the maximum at about Day 30.On the contrary, when they are in combined use, the tumor growth was significantly delayed and the effect of growth reverse was also observed as shown in Figure 19.
Example 20: Orin1001 inhibits non-small cell lung cancer in combination  with paclitaxel
Orin1001 was administered by oral gavage in a xenograft mouse model using female NCr nu/nu mice injected subcutaneously with A549 human lung carcinoma tumor cells. Animals were assigned to 4 groups (n=10/group) ; Vehicle control administered by oral gavage for 28 days, Orin1001 administered via oral gavage at 300 mg/kg/day for 28 consecutive days, paclitaxel administered weekly by iv at 15 mg/kg, or Orin1001 administered in combination with paclitaxel. The results are shown in Figure 20.
As shown in Figure 20, in combination with paclitaxel, when Orin1001 was used together with paclitaxel, it showed a modest increase in tumor inhibition compared to ORIN1001 or paclitaxel alone.
Example 21: Orin1001 inhibits liver cancer in combination with sorafenib
The inhibiting effects of Orin1001 against liver cancer growth in combination with sorafenib was tested in a subcutaneous Hep3B (ATCC, Manassas, VA, cat #HB-8064) human liver xenograft  model with female BALB/c-Nu/Nu mice. Animals were assigned to 6 groups (n=10/group) . Sorafenib was purchased from Bide Pharmatech LTD and formulated as a solution 
Figure PCTCN2018096613-appb-000058
EL/ethanol (50: 50) . The results shown in Figure 21 indicated an interim result that Orin1001 synergistically inhibits Hep3B tumor xenografts in combination with sorafenib. Although the study was still ongoing, the 8 th day data already showed the synergistic effect.
Example 22: Orin1001 was chosen to be used in combination with other therapeutic agents Example 22 was related to comparative PD/PK data of Orin 1001 VS compound 4315, as its structure shown below, which is an earlier lead of this series of compounds. Tunicamycin was used herein to activate IRE1, then Orin1001 (also called 4485) or 4315 were given to inhibit the activating effect. Orin1001 demonstrated potent in vivo potency inhibiting IRE1α in our liver PD screening assay. The results are shown in Figure 22. Each gel panel represents one mouse liver sample as in the figure.
BALB/c mice was injected intraperitoneally with 100 microliters at an equivalent dose of 1 mg/kg tunicamycin solution. Two hours after tunicamycin injection, mice were dosed with compound of interest either PO or IV. Following 2 hours for PO delivery of compound, mice were euthanized according to IACUC protocols using CO 2 from a compressed air source. A 1 cm 3 fragment of the liver for homogenization and extraction of the RNA were collected for further analysis. Total RNA is harvested from cells or tissue using TRIzol according to the manufacturer’s procedures. After ethanol precipitation and resuspension of the RNA, RiboGreen (Invitrogen) is used to quantify the yield and normalize the RNA concentration in the source tube containing isolated RNA. RT-PCR is performed by Oligo (dT) priming, and SuperScript II (Invitrogen) transcription using the Amplitaq Gold Kit (Applied Biosystems) according to the manufacturer’s protocols. Primers for human XBP-1 are 5_-CCTGGTTGCTGAAGAGGAGG-3_ (forward, Seq ID No. 8) and 5_-CCATGGGGAGATGTTCTGGAG-3_ (reverse, Seq ID No. 9) , and for mouse are 5_-ACACGCTTGGGAATGGACAC-3_ (forward, Seq ID No. 10) and 5_-CCATGGGAAGATGTTCTGGG-3 (Seq ID No. 11) . All DNA oligos were purchased from IDT DNA Technologies. PCR is run on a Bio-Rad PTC-100 96-well thermocycler with heating at 94 ℃ for 30 s, annealing at 58 ℃ for 30 s, and polymerizing at 72 ℃ for 30 s for 35 cycles. Reactions are run on 4% precast NuSieve gels from Cambrex and visualized by ethidium bromide staining and UV excitation.
Accuprime kit (12339-024, Invitrogen) 
mXBP-1 458 5’ -GAGGCCAAGGGGAGTGGA-3’ (custom order, IDT) (Seq ID No. 12) 
mXBP-1 572 5’ -AGATGTTCTGGGGAGGTGACAACT-3’ (custom order, IDT) (Seq ID No. 13) mGAPDH 548 (custom order, IDT)
mXBP-1 524 UnSp 5’ Tex-CACATAGTCTGAGTGTGCTG-3’ BHQ-2 (custom order, Biosearch Technologies) (Seq ID No. 14) 
mXBP-1 580 Sp 5’ FAM-CCTGCACCTGCTGCGGACT-3’ BHQ-1 (custom order, Biosearch Technologies) (Seq ID No. 15) 
mGAPDH 608 5’ HEX/3’ BHQ-1 (custom order, Biosearch Technologies) yeast tRNA (54016, Invitrogen) thin-wall 96 well RTq PCR plate.
All PK experiments are standard tests that performed in either WuXi PharmaTech or Charels River Laboratory. 4315 and 4485 (Orin1001) were dosed PO as a suspnesion in 1% microcellulose (Sigma) and 50% Sucrouse (Sigma) .
As shown in Figure 22, as an example, Orin1001 has an ED 50 less than 2 mg/kg PO vs 4315 which has an ED 50 >10 mg/kg. 4315 ED 50 was determined to be 50 mg/kg in a separate experiment. Compound 4315 is disclosed in WO2011/127070 A2 as compound B, a preferred IRE1α inhibitor. In the figure, top panel when labeled as PBS/4315 or Tun/4315, they meant the mice was either dosed with PBS buffer or tunicamycin to active the IRE1α so that XBP1s was observed as illustrated in the lower panel. In the middle panel, all the 4485 dosing group mice were dosed firstly with tunicamycin.
Orin1001 (4485) also has much improved oral bioavilability as shown in the table cross all tested spices.
Figure PCTCN2018096613-appb-000059
  4315 oral bioavailability 4485 (Orin1001) oral bioavilability
Mouse 23% (5mg/kg dose) 63% (10mg/kg dose)
Rat 36% (10mg/kg dose) 52% (10mg/kg dose)
Dog 17% (10mg/kg dose) 68.5% (4mg/kg dose)

Claims (22)

  1. A pharmaceutical combination, comprising
    (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof, and (b) one or more additional cancer therapeutic agents:
    Figure PCTCN2018096613-appb-100001
    wherein
    R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1-C 6 hydrocarbon chain containing N or O atom, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
    R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino;
    R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6 alkoxyl, C 1-6 hydroxylalkyl, C 1-6 alkoxylC 1-6alkyl, 
    Figure PCTCN2018096613-appb-100002
    Figure PCTCN2018096613-appb-100003
    R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6alkoxyl C 1-6alkyl; or
    R 9 and R 10 together with the nitrogen atom to which they are attached form a heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6 alkylamino, C 1-6 alkoxyl.
  2. The pharmaceutical combination according to claim 1, wherein
    the additional cancer therapeutic agent has at least one of the following features:
    (1) inducing ER stress;
    (2) inducing or up-regulating IRE-1α expression;
    (3) inducing or up-regulating XBP1 splicing; and
    (4) being less effective when IRE-1α is expressed.
  3. The pharmaceutical combination according to claim 1 or 2, wherein the compound of formula  (I) or the pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents are administered simultaneously, separately or sequentially.
  4. The pharmaceutical combination according to any one of claims 1-3, wherein
    the compound of formula (I) has formula (II)
    Figure PCTCN2018096613-appb-100004
  5. The pharmaceutical combination according to any one of claims 1-4, wherein
    the pharmaceutical combination is in the form of a pharmaceutical composition or a kit.
  6. The pharmaceutical combination according to claim 1, wherein
    the additional cancer therapeutic agent is selected from the group consisting of
    cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents; immunosuppressives; anti-angiogenic compounds and vascular endothelial growth factor inhibitors; fibroblast growth factor inhibitors; epidermal growth factor receptor inhibitors; antibodies; checkpoint inhibitors; PD-1 inhibitors or PD-L1 inhibitors; cell cycle inhibitors and differentiation inducers; mTOR inhibitors; corticosteroids; growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers; caspase activators; chromatin disruptors and DNA repair enzyme inhibitors; HDAC inhibitors; Bcr-Abl inhibitors; FMS-like tyrosine kinase 3 (Flt3) inhibitors; and preferably selected from the group consisting of lestaurtinib, nilotinib, sorafenib, dasatinib, gefitinib, temisirolimus, vatalinib, 
    Figure PCTCN2018096613-appb-100005
    vorinostat, paclitaxel, gemcitabine, 17-AAG, 
    Figure PCTCN2018096613-appb-100006
    tamoxifen, more preferably selected from the group consisting of paclitaxel, 
    Figure PCTCN2018096613-appb-100007
    and tamoxifen.
  7. A kit or a pharmaceutical composition, comprising the pharmaceutical combination according to any one of claims 1-6.
  8. A method for treating cancerous tumor, comprising administering a subject in need thereof an effective amount of the pharmaceutical combination according to any one of claims 1-6 or the kit or pharmaceutical composition according to claim 7.
  9. The method according to claim 8, wherein the cancerous tumor is selected from the group consisting of liver cancer, triple negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma and multiple myeloma.
  10. The method according to claim 8 or 9, wherein the compound of formula (I) or the pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents are administered simultaneously, separately or sequentially.
  11. A method for enhancing the efficacy of a cancer therapeutic agent, comprising applying the compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with the cancer therapeutic agent,
    Figure PCTCN2018096613-appb-100008
    wherein
    R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1-C 6 hydrocarbon chain containing N or O atom, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
    R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino;
    R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6 alkoxyl, C 1-6 hydroxylalkyl, C 1-6 alkoxylC 1-6alkyl, 
    Figure PCTCN2018096613-appb-100009
    Figure PCTCN2018096613-appb-100010
    R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6alkoxyl C 1-6alkyl; or
    R 9 and R 10 together with the nitrogen atom to which they are attached form a heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6 alkylamino, C 1-6 alkoxyl.
  12. The method according to claim 11, wherein the cancer therapeutic agent has at least one of  the following features:
    (1) inducing ER stress;
    (2) inducing or up-regulating IRE-1α expression;
    (3) inducing or up-regulating XBP1 splicing; and
    (4) being less effective when IRE-1α is expressed.
  13. The method according to claim 11 or 12, wherein the compound of formula (I) or the pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents are administered simultaneously, separately or sequentially.
  14. The method according to any one of claims 11-13, wherein
    the compound of formula (I) has formula (II)
    Figure PCTCN2018096613-appb-100011
  15. The method according to any one of claims 11-14, wherein the cancer therapeutic agent is selected from the group consisting of cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents; immunosuppressives; anti-angiogenic compounds and vascular endothelial growth factor inhibitors; fibroblast growth factor inhibitors; epidermal growth factor receptor inhibitors; antibodies; checkpoint inhibitors; PD-1 inhibitors or PD-L1 inhibitors; cell cycle inhibitors and differentiation inducers; mTOR inhibitors; corticosteroids; growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers; caspase activators; chromatin disruptors and DNA repair enzyme inhibitors; HDAC inhibitors; Bcr-Abl inhibitors; FMS-like tyrosine kinase 3 (Flt3) inhibitors; and preferably selected from the group consisting of lestaurtinib, nilotinib, sorafenib, dasatinib, gefitinib, temisirolimus, vatalinib, 
    Figure PCTCN2018096613-appb-100012
    vorinostat, paclitaxel, gemcitabine, 17-AAG, 
    Figure PCTCN2018096613-appb-100013
    tamoxifen, more preferably selected from the group consisting of paclitaxel, 
    Figure PCTCN2018096613-appb-100014
    and tamoxifen.
  16. The method according to any one of claims 11-15, wherein the cancer therapeutic agent is used for treatment of cancerous tumor selected from the group consisting of liver cancer, triple  negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma and multiple myeloma.
  17. A method for treating cancerous tumors, comprising administering a subject in need thereof an effective amount of formula (I) or a pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents:
    Figure PCTCN2018096613-appb-100015
    wherein
    R 3 and R 4 are independently hydrogen or C 1-6 alkoxyl, which is optionally substituted with one or more substituents selected from the group consisting of (1) C 1-C 6 hydrocarbon chain containing N or O atom, and (2) C 3-10 cycloalkyl, which optionally contains 1 or 2 heteroatoms independently selected from the group consisting of N, O, and S;
    R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxyl, or C 1-6 alkylamino;
    R 6 is C 1-6 alkyl, which is substituted with 1, 2 or 3 substituents independently selected from the group consisting of C 1-6 alkoxyl, C 1-6 hydroxylalkyl, C 1-6 alkoxylC 1-6alkyl, 
    Figure PCTCN2018096613-appb-100016
    Figure PCTCN2018096613-appb-100017
    R 9 and R 10 are independently hydrogen; C 1-6 alkyl; C 1-6 alkoxyl C 1-6 alkyl; perfluoro C 1-6alkoxyl C 1-6alkyl; or
    R 9 and R 10 together with the nitrogen atom to which they are attached form a heterocycle containing 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S, and the heterocycle is optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 1-6 alkylamino, C 1-6 alkoxyl.
  18. The method according to claim 17, wherein the cancer therapeutic agent has at least one of the following features:
    (1) inducing ER stress;
    (2) inducing or up-regulating IRE-1α expression;
    (3) inducing or up-regulating XBP1 splicing; and
    (4) being less effective when IRE-1α is expressed.
  19. The method according to claim 17 or 18, wherein the compound of formula (I) or the pharmaceutically acceptable salt thereof and one or more additional cancer therapeutic agents are administered simultaneously, separately or sequentially.
  20. The method according to any one of clams 17-19, wherein the compound of formula (I) has formula (II)
    Figure PCTCN2018096613-appb-100018
  21. The method according to any one of claims 17-20, wherein the cancer therapeutic agent is selected from the group consisting of cytotoxic chemotherapeutic agents; antimetabolites; antimitotic agents; alkylating agents; DNA damaging agents; antitumor antibiotics; platinum coordination complexes; proteasome inhibitors; HSP90 inhibitors; hormones and hormone analogs; aromatase inhibitors; fibrinolytic agents; antimigratory agents; antisecretory agents; immunosuppressives; anti-angiogenic compounds and vascular endothelial growth factor inhibitors; fibroblast growth factor inhibitors; epidermal growth factor receptor inhibitors; antibodies; checkpoint inhibitors; PD-1 inhibitors or PD-L1 inhibitors; cell cycle inhibitors and differentiation inducers; mTOR inhibitors; corticosteroids; growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers; caspase activators; chromatin disruptors and DNA repair enzyme inhibitors; HDAC inhibitors; Bcr-Abl inhibitors; FMS-like tyrosine kinase 3 (Flt3) inhibitors; and preferably selected from the group consisting of lestaurtinib, nilotinib, sorafenib, dasatinib, gefitinib, temisirolimus, vatalinib, 
    Figure PCTCN2018096613-appb-100019
    vorinostat, paclitaxel, gemcitabine, 17-AAG, 
    Figure PCTCN2018096613-appb-100020
    tamoxifen, more preferably selected from the group consisting of paclitaxel, 
    Figure PCTCN2018096613-appb-100021
    and tamoxifen.
  22. The method of according to any one of claims 17-21, wherein the cancerous tumor is selected from the group consisting of liver cancer, triple negative breast cancer, estrogen positive breast cancer, ovarian carcinoma, pancreatic cancer, head and neck cancer, non-small cell lung cancer, glioblastoma and multiple myeloma.
PCT/CN2018/096613 2018-07-23 2018-07-23 IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT WO2020019107A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
PCT/CN2018/096613 WO2020019107A1 (en) 2018-07-23 2018-07-23 IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT
KR1020217004949A KR20210036374A (en) 2018-07-23 2019-07-23 IRE1α inhibitors in combination with cancer treatments for cancer treatment
CN201980063917.7A CN113164457A (en) 2018-07-23 2019-07-23 IRE1 alpha inhibitors in combination with cancer therapeutics for cancer treatment
PCT/CN2019/097291 WO2020020155A1 (en) 2018-07-23 2019-07-23 IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT
AU2019311031A AU2019311031A1 (en) 2018-07-23 2019-07-23 IRE1α inhibitor in combination with cancer therapeutic agent for cancer treatment
US17/261,413 US20210260069A1 (en) 2018-07-23 2019-07-23 IRE1a INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT
EP19840929.4A EP3826634A4 (en) 2018-07-23 2019-07-23 Ire1 alpha inhibitor in combination with cancer therapeutic agent for cancer treatment
JP2021503583A JP7468829B2 (en) 2018-07-23 2019-07-23 IRE1α INHIBITORS IN COMBINATION WITH CANCER THERAPEUTICS FOR TREATING CANCER - Patent application
CA3106731A CA3106731A1 (en) 2018-07-23 2019-07-23 Ire1a inhibitor in combination with cancer therapeutic agent for cancer treatment

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2018/096613 WO2020019107A1 (en) 2018-07-23 2018-07-23 IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT

Publications (1)

Publication Number Publication Date
WO2020019107A1 true WO2020019107A1 (en) 2020-01-30

Family

ID=69180852

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/096613 WO2020019107A1 (en) 2018-07-23 2018-07-23 IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT

Country Status (1)

Country Link
WO (1) WO2020019107A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022106579A1 (en) * 2020-11-20 2022-05-27 Institut National De La Sante Et De La Recherche Medicale (Inserm) Compounds for treating a disease associated with macrophage senescence

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102015606A (en) * 2007-06-08 2011-04-13 满康德股份有限公司 IRE-1a inhibitors
CN103079558A (en) * 2010-04-05 2013-05-01 满康德股份有限公司 IRE-1[alpha] inhibitors
CN106822904A (en) * 2017-03-08 2017-06-13 暨南大学 The pharmaceutical composition and its application of inhibitor containing AKT and IRE1 inhibitor
CN106822905A (en) * 2017-03-08 2017-06-13 暨南大学 The medicine and purposes of inhibitor containing Survivin and IRE1 inhibitor
CN106974908A (en) * 2017-03-02 2017-07-25 深圳大学 Pharmaceutical composition and purposes containing hdac inhibitor and IRE1 inhibitor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102015606A (en) * 2007-06-08 2011-04-13 满康德股份有限公司 IRE-1a inhibitors
CN103079558A (en) * 2010-04-05 2013-05-01 满康德股份有限公司 IRE-1[alpha] inhibitors
CN106974908A (en) * 2017-03-02 2017-07-25 深圳大学 Pharmaceutical composition and purposes containing hdac inhibitor and IRE1 inhibitor
CN106822904A (en) * 2017-03-08 2017-06-13 暨南大学 The pharmaceutical composition and its application of inhibitor containing AKT and IRE1 inhibitor
CN106822905A (en) * 2017-03-08 2017-06-13 暨南大学 The medicine and purposes of inhibitor containing Survivin and IRE1 inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MING J. ET AL.: "A novel chemical, STF-083010, reverses tamoxifen-related drug resistance in breast cancer by inhibiting IRE1/XBP1", ONCOTARGET, vol. 6, no. 38, 1 December 2015 (2015-12-01), pages 40692 - 40703, XP055557077 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022106579A1 (en) * 2020-11-20 2022-05-27 Institut National De La Sante Et De La Recherche Medicale (Inserm) Compounds for treating a disease associated with macrophage senescence

Similar Documents

Publication Publication Date Title
AU2006210746B2 (en) Combination therapies using HDAC inhibitors
CA2782555C (en) 3-(indolyl)-or 3-(azaindolyl)-4-arylmaleimide derivatives for use in the treatment of colon and gastric adenocarcinoma
RU2731908C2 (en) Apilimod composition and methods of using it in treating melanoma
JP2017503842A (en) Apilimod composition for cancer treatment
TWI685341B (en) Use of combination of apatinib and c-met inhibitor in the preparation of medicament for treating tumor
KR20200096788A (en) Use of PARP inhibitors in chemotherapy-resistant ovarian or breast cancer treatment
CA2982435A1 (en) Treatment method by combined use of mdm2 inhibitor and btk inhibitor
WO2020019107A1 (en) IRE1α INHIBITOR IN COMBINATION WITH CANCER THERAPEUTIC AGENT FOR CANCER TREATMENT
JP7468829B2 (en) IRE1α INHIBITORS IN COMBINATION WITH CANCER THERAPEUTICS FOR TREATING CANCER - Patent application
WO2020087522A1 (en) IRE1α inhibitor in combination with cancer therapeutic agent for cancer treatment
CA3121441C (en) Ezh1/2 dual inhibitor-containing pharmaceutical composition to be used as a combination drug
WO2018213770A1 (en) Therapeutic methods
WO2021110136A1 (en) Pharmaceutical combination and use thereof
US20220047573A1 (en) Methods for treating vascular malformations
WO2021119397A9 (en) Metal salts and uses thereof
WO2022171064A1 (en) Pharmaceutical use of nicotinamide and composition containing same
TWI837231B (en) Pharmaceutical combination containing ezh1/2 dual inhibitor and use thereof
WO2021030404A1 (en) Methods and compositions for treating vascular malformations
WO2023168491A1 (en) Treatment of clear cell renal cell carcinoma
AU2012202000B2 (en) Combination therapies using HDAC inhibitors

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: 18928134

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18928134

Country of ref document: EP

Kind code of ref document: A1