WO2018098352A2 - Ciblage d'expression du point de contrôle immunitaire induit par kras - Google Patents

Ciblage d'expression du point de contrôle immunitaire induit par kras Download PDF

Info

Publication number
WO2018098352A2
WO2018098352A2 PCT/US2017/063116 US2017063116W WO2018098352A2 WO 2018098352 A2 WO2018098352 A2 WO 2018098352A2 US 2017063116 W US2017063116 W US 2017063116W WO 2018098352 A2 WO2018098352 A2 WO 2018098352A2
Authority
WO
WIPO (PCT)
Prior art keywords
kras
certain embodiments
modulator
signaling
cancer
Prior art date
Application number
PCT/US2017/063116
Other languages
English (en)
Other versions
WO2018098352A3 (fr
Inventor
Jun Oishi
Xiangao Sun
Chiang J. Li
Original Assignee
Jun Oishi
Xiangao Sun
Li Chiang J
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 Jun Oishi, Xiangao Sun, Li Chiang J filed Critical Jun Oishi
Publication of WO2018098352A2 publication Critical patent/WO2018098352A2/fr
Publication of WO2018098352A3 publication Critical patent/WO2018098352A3/fr

Links

Classifications

    • 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
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • 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
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles 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
    • A61K31/435Heterocyclic 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
    • A61K31/437Heterocyclic 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 five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • cytotoxic T lymphocytes can orchestrate potent anti-tumor immune responses involving both adaptive and innate effector mechanisms. Nonetheless, tumor cells are often able to evade this immune surveillance by commandeering immune checkpoint inhibitory pathways that are hardwired into the immune system to retain self-tolerance and modulate the duration and amplitude of physiological immune responses in order to minimize potential collateral tissue damage.
  • the PD-1 immune checkpoint pathway is one such example of an immune checkpoint that has emerged as a critical mediator of immunosuppression in the local tumor microenvironment.
  • the inliibitory co-receptor Programmed Death 1 (PD-1; also known as CD279), a member of the extended CD28/CTLA-4 family of T cell regulators, is expressed on immune cells, such as T, B and NIC cells, whereas its ligand, the Programmed Cell Death Ligand 1 (PD-L1 , also known as CD274 or B7-H1) is a cell surface glycoprotein expressed on the surface of tumor cells of solid tumors as well as on human tumor associated antigen presenting cells (APCs), e.g., dendritic cells and macrophages.
  • APCs human tumor associated antigen presenting cells
  • the interaction of PD-L 1 ligand on tumor cells with the PD-1 receptor on immune cells delivers an inliibitory signal to T lymphocytes that ultimately leads to T cell anergy and immune evasion.
  • the present disclosure is based on the discovery that aberrant KRAS signaling is at least in part responsible for the activation of PD-L 1 gene expression in tumor ceils and for the subsequent suppression of tumor cell-specific T cell toxicity.
  • the present disclosure provides compositions and methods that can prevent tumors from evading immune surveillance through Lhe aberrant activation of the PD-Ll/PD-1 immune checkpoint pathway in T cells, in certain embodiments, inhibition of abeiTant KRAS signaling in tumor cells sensitizes tumor cells to immune checkpoint inhibitors.
  • the disclosure further provides methods for enhancing the therapeutic efficacy of existing anticancer treatment using KRAS signaling modulators.
  • a composition comprising an effective amount of a modulator of KRAS signaling, wherein the modulator of KRAS signaling is effective at enhancing the sensitivity of a tumor cell to tumor cell-specific T cell cytotoxicity.
  • the tumor cell is in a subject, in certain embodiments, the modulator of KRAS signaling enhances the efficacy of a therapeutic agent at treating a KRAS associated disease, e.g. cancer.
  • the modulator of KRAS signaling can be, for example, an inhibitor of aberrant KRAS signaling.
  • the abeiTant KRAS signaling is induced by a modified KRAS, e.g., an oncogenic KRAS, expressed in tumor cells.
  • the aberrant KR AS signaling comprises the KRAS induced activation of at least one member of the RAS/R AF/ME /'ERK/FR A ⁇ 1 signal transduction pathway, in certain embodiments, the abeiTant KRAS signaling can result irs the KRAS induced activation of PD- L 1 gene expression in tumor cells.
  • the aberrant. KRAS signal ing cars be induced by an effector of KRAS signaling, e.g. by a KRAS GEF.
  • a composition in a second aspect, comprises a combination of an effective amount of a modulator of KRAS signaling, and an effective amount of a therapeutic agent, wherein the modulator of KRAS signaling is effective at enhancing the sensitivity of a tumor cell to tumor cell-specific T cell cytotoxicity.
  • the modulator of KRAS signaling can be an inhibitor ofKRAS signaling.
  • the modulator of KRAS signaling can enhance the efficacy of the therapeutic agent at treating a KRAS associated disease, for example, cancer.
  • the tumor cell is in a subj ect.
  • a composition in a third aspect, comprises a combination of an effective amount of an modulator of oncogenic KRAS signaling, and an effective amount of a therapeutic agent, wherein the modulator of oncogeni c KRAS signaling is effective at enhancing the sensitivity of a tumor cell to tumor cell-specific T cell cytotoxicity.
  • the modulator of oncogenic KRAS signaling can be an inhibitor of oncogenic KRAS signaling.
  • the modulator of oncogenic KRAS signaling can enhance die efficacy of the therapeutic agent at treating an oncogenic KR AS associated disease, in certain embodiments, the tumor cell is in a subject.
  • the therapeutic agent can be, for example, an anticancer therapeutic agent.
  • the anticancer therapeutic agent can be, for example, an immunotherapeutic agent, such as an antigen-binding protein, or fragment thereof, e.g. an antibody that targets a cell surface antigen or extracellular growth factor.
  • the anticancer therapeutic agent can be, for example, a small molecule inhibitor of a target protein required for the maintenance or progression of a cancer.
  • the small molecule inhibitor can be a small molecule proteasome inhibitor, a small molecule tyrosine kinase inhibitor, a small molecule cyclin-dependent kinase inhibitor, a small molecule inliibitor of a transcription factor or s small molecule inhibitor of an immune checkpoint molecule.
  • the anticancer therapeutic agent can be, for example, an RNA interfering agent that silences the expression of a target gene required for the maintenance or progression of a cancer.
  • the therapeutic agent can be, for example, an epigenetic inhibitor, e.g. an HDAC inhibitor.
  • the anticancer therapeutic agent can be, for example, an anti-estrogen or an anti-androgen therapeutic agent.
  • the anticancer therapeutic agent can be, for example, a chemotherapeutic agent arid/or radiotherapy.
  • the anticancer therapeutic agent can be, for example, a cancer vaccine.
  • the anticancer therapeutic agent can be, for example, an RNA interfering agent of an immune checkpoint molecule, e.g. PD-L1.
  • a composition in a fourth aspect, comprises a combination of an effective amount of a modulator of KRAS signaling, and an effective amount of an immune checkpoint inliibitor, wherein the modulator of KRAS signaling is effective at enhancing the sensitivity of a tumor cell to the immune checkpoint inliibitor.
  • the tumor cells can be resistant or have acquired resistance to the immune checkpoint inliibitor.
  • the modulator of KRAS signaling sensitizes a tumor cell to tumor cell - specific T cell cj' otoxicity.
  • the modulator of KRAS signaling enhances the efficacy of the immune checkpoint inhibitor at treating a KRAS associated disease, e.g., cancer.
  • the tumor cell is in a subject.
  • a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling and an effective amount of an immune checkpoint inhibitor to the subject with the cancer, wherein the administration of the modulator of KRAS signaling and the immune checkpoint inhibitor is effective at enhancing the sensitivity of the tumor cell s to the immune checkpoint inhibitor.
  • the tumor cell is in a subject.
  • the modulator of KRAS signaling can act in synergy with the immune checkpoint inhibitor to enhance an immune response against a tumor.
  • the immune checkpoint inhibitor is effective at blocking the interaction of programmed cell death protein 1 (PD-1) receptor with programmed cell death 1 ligand 1 (RD-L1).
  • the immune checkpoint inhibitor can be, for example, ipilimumab, tremelimumab, atezolizumab, nivolumab, pembrolizumab, JS001, REGN2810, SHR- 121Q, MEDI0680, FDR001, BGB-A317, TSR-042, PF-06801591, Ningbo Cancer Hosp. anti-PD-I CAR, Medimmune anti-PD- l , s anti-PD- i. UCB anti-PD-l or 948.
  • the mmune checkpoint inhibitor is effective at inhibiting an endogenous immune checkpoint protein or fragment thereof chosen from, for example, PD-1, PD-L1, PD-L2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7 ⁇ H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3.
  • the tumor cells express a modified KRAS, e.g. an oncogenic KRAS.
  • the expressed oncogenic KRAS comprises a mutation of at least one amino acid residue of the amino acid sequence of SEQ ID No. :980.
  • the mutation can be an activating mutation of KRAS.
  • the oncogenic KRAS comprises an activating mutation of amino acid residues G12, G13, S17, P34 and/or Q61 of SEQ ID No.; 980.
  • the oncogenic KRAS comprises a mutation chosen from G12C, G12S, G 12R, G12F, G12L, G12N, G12A, G12D, GOV, G13C, G13S, G1.3D, G13V, G13P, S17G, P34S, Q61K, Q61L, Q61R, and/or Q61H.
  • the modulator of KRAS signaling inhibits aberrant KRAS signaling.
  • the aberrant KRAS signaling comprises signaling by an oncogenic KRAS expressed in the tumor cells.
  • the aberrant KRAS signaling comprises the KRAS induced activation of at least one effector of the RAS/ RAF MEK'' ERK ' FRA-1 signal transduction pathway in tumor cells.
  • the aberrant KRAS signaling comprises the KRAS induced activation of PD-L1 gene expression in tumor ceils.
  • the modulator of KRAS signaling is ineffective at reducing KRAS induced signaling activity in the absence of oncogenic KRAS gene expression.
  • the modulator of KRAS signaling can be effective at inhibiting the level of KRAS mRNA in the tumor cells by at least about 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, by at least about 80%, by at least about 90% by at least about. 93% or by at least about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the level of KRAS mRNA in the tumor cells by at least 95%.
  • the modulator of KRAS signaling can be effective at inhibiting the level of KRAS mRNA in the tumor cells by at least. 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the level of KRAS mRNA in the tumor cells from about 10% to about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the level of KRAS induced PD-L1 gene expression in the tumor cells by at least about 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%. by at least about 60%, by at least about 70%, or by at least about 75% by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95% or by at least about 99%.
  • the modulator of KRAS signaling can be effective at mhibiting the level of KRAS induced PD-L1 gene expression in the tumor cells by at least about 80%, in certain embodiments, the modulator of KR AS signaling can be effective at inhibiting the level of KRAS induced PD-L1 gene expression in the tumor cells from about 10% to about 99%
  • the modulator of KRAS signaling can be effective at inhibiting the level of FRA-1 gene expression in the tumor cells by at least about. 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least, about 60% by at least about 70%, or by at least about 75% by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95% or by at least about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the level of FRA-1 gene expression in the tumor cells by at least about 80%.
  • the modulator of KRAS signaling can be effective at inhibiting the level of FRA-1 gene expression in the tumor cells from about 10% to about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the KRAS induced activation of at least one effector molecule of the RAS/ RAF/ MEK/ ER ' FRA- 1 signal transduction pathway in tumor cells.
  • the modulator of KRAS signaling can be effective at inhibiting the KRAS induced activation of at least one effector molecule of the RAS/ RAF/ MEK/ ERK/ FRA- 1 signal transduction pathway in tumor by at least about 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, or by at least about 75% by at least about 80% by at least about 85%, by at least about 90%, by at least about 95% or by at least about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the KRAS induced activation of at least one effector molecule of the RAS/ RAF/ MEK/ ERK/ FRA- 1 signal tra nsduction pathway in tumor from about 10% to about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the KRAS-induced phosphorylation of RAF, MEK. or ERK in tumor cells.
  • the modulator of KRAS signaling cart be effective at inhibiting the KRAS-induced phosphorylation of RAF, MEK or ERK in tumor cells by at least about 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, or by at least about 75% by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95% or by at least, about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting the KRAS-induced phosphorylation of RAF, MEK or ERK in tumor cells from about 10% to about 99%.
  • the modulator of KRA.S signaling can be effective at inhibiting the level of both KRAS and PD-L1 gene expression in tumor cells by at least about 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, or by at least about 75% by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95% or by at least about 99%.
  • the modulator of KRAS signaling can be effective at inhibiting both KRAS and PD-L1 gene expression in tumor cel ls from about 10% to about 5)9%.
  • the modulator of KRAS signaling can be effective at inhibiting KRAS, RAF. IvIEK, ERK, FRA-1 and PD-L1 signaling activity in tumor cells from about 10% to about 99%.
  • the modulator of KRAS signaling comprises an RNA interfering agent.
  • the RNA interfering agent targets the expression of one or more effectors of the RAS/ RAF MEK7 ERK' FRA-1 signal transduction pathway.
  • the modulator of KRAS signaling comprises an inhibitor of GTP bound KRAS activity. In certain embodiments, the modulator of KRAS signaling comprises an inhibitor of a KRAS GEF. in certain embod ments, the modulator of KRAS signaling comprises an activator of KRAS bound GTP hydrolysis, e.g. KRAS GAP activity. In certain embodiments, the modulator of KRAS signaling comprises an inhibitor of oncogenic KRAS. In certain embodiments, the modulator of KRAS signaling comprises a KRAS-specifie RNA interfering agent, e.g. a KRAS-specific asymmetric interfering RNA (referred to herein as KRAS aiRNA or aiKRAS).
  • KRAS aiRNA or aiKRAS KRAS-specific asymmetric interfering RNA
  • the modulator of KRAS signaling comprises an oncogenic KRAS-specific RNA interfering agent, e.g. an oncogenic KRAS- specific asymmetric interfering RNA.
  • the modulator of KRAS si gnaling comprises an RNA interfering agent that targets both wild type and oncogenic KRAS, e.g. a KRAS-specific asymmetric interferin RNA.
  • the tumor can be, for example, a tumor caused by pancreatic ductal adenocarcinoma (PDAC), colorectal cancer, or non-small-cell lung cancer (NSCLC).
  • PDAC pancreatic ductal adenocarcinoma
  • NSCLC non-small-cell lung cancer
  • the cancer can be a metastatic cancer, a cancer that is refractory to chemotherapy, a cancer that is refractory to radiotherapy and/or a cancer that has relapsed.
  • the cancer can be resistant to an immunotherapeutic agent, e.g. an immune checkpoint inhibitor.
  • the disclosure provides a modulator of oncogenic KRAS signaling.
  • the oncogenic KRAS signaling comprises aberrant KRAS signaling.
  • the modulator of KRAS signaling is effective at inhibiting oncogenic KRAS signaling.
  • the modulator of KRAS signaling is ineffective at reducing
  • the modulator of KRAS signaling comprises an RNA interfering agent, for example, an asymmetric interfering RNA (aiRNA).
  • aiRNA asymmetric interfering RNA
  • the asymmetric interfering RNA comprises a sense strand sequence that is at least 50% identical to a sequence chosen from SEQ ID NOs: 320-637.
  • the asymmetric interfering RNA comprises a sense strand sequence chosen from SEQ ID NOs: 320-637.
  • the asymmetric interfering RNA comprises an antisense strand sequence that is at least 50% identical to a sequence chosen from SEQ ID NOs: 638-955.
  • the asymmetric interfering RNA comprises an antisense strand sequence chosen from SEQ ID NOs: 638-955.
  • the disclosure provides a composition comprising an effective amount of a modulator of oncogenic KRAS signaling.
  • the disclosure provides a method for changing the efficacy or/and safety of a therapeutic agent comprising administering an effective amount of a modulator of KRAS signaling.
  • the modulator of KRAS signaling can act in synergy with the therapeutic agent to enhance the efficacy and/or safety of the therapeutic agent at treating cancer.
  • the disclosure provides a method for changing the efficacy or/and safety of a therapeutic agent comprising administering an effective amount of an asymmetric interfering RNA (aiRNA).
  • aiRNA asymmetric interfering RNA
  • the asymmetric interfering RNA (aiRNA) can act in synergy with the therapeutic agent to enhance the efficacy and/or safety of the therapeutic agent at treating cancer.
  • the disclosure provides a method for changing the efficacy or/and safety of a therapeutic agent comprising administering an effective amount of an asymmetric
  • S interfering RNA comprising a sense strand sequence chosen from SEQ ID.NOs: 320- 637.
  • the disclosure provides a method for changing the efficacy or/and safety of a therapeutic agent comprising administering an effective amount of an asymmetric interfering RNA (aiRN A) comprising an antisense strand sequence chosen from SEQ ID NOs: 638-955.
  • aiRN A asymmetric interfering RNA
  • the efficacy of the therapeutic agent is enhanced
  • the safety of the therapeutic agent is enhanced
  • the therapeutic agent is an immune checkpoint inhibitor.
  • the therapeutic agent is chosen, for example, from ipilimumab, trerneiimumab, atezolizumab, nivolumab, pembrolizumab, JS001, REGN2810, SHR-1210, MEDIQ68Q, PDR001, BGB-A317, TSR-042, PF-06801591, Ningbo Cancer Hosp.
  • anti-PD- 1 CAR edimmune anti-PD-1 5 Isis anti-PD-1, UCB anti-PD-l or 948,gl, Dana-Farber anti-PD- 1 , STT-1 1 10, Suzhou Stainwei Biotech anti-PD- 1 , Haixi pembrolizumab btosimilar, Livzon anti-PD- 1, MabQuest anti-PD-1, Singapore ASTR.
  • the therapeutic agent can be effective at inhibiting an endogenous immune checkpoint protein or fragment thereof chosen from, for example, PD-1 , PD-L1, PD-L2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7- H3, B7-H4, BTLA HVEM, KIR, TCR, LAG3, CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, ⁇ 3, GAL9, ADORA CD276, VTCN1, IDOl, KJR3DL1, HAVCR2, VISTA and/or CD244 or any combination thereof.
  • an endogenous immune checkpoint protein or fragment thereof chosen from, for example, PD-1 , PD-L1, PD-L2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7- H3, B7-H4, BTLA HVEM, KIR, TCR, LAG3, CD
  • Fig. ⁇ A shows the nucleotide sequences within the KRAS gene that are targeted as well as the sequences of the sense and antisense strands of the scrambled aiControi and exemplar)'. interfering RNAs aiKRAS#l, aiKRAS#2, aiKRAS#3, aiKRAS#4, aiPD-Ll, aiFra-l#l, and aiFra-l#2.
  • FIG. IB depicts an exemplary nucleotide sequence of the human KRAS proto- oncogene, transcript variant B mRNA (NCBI Reference Sequence: NM 004985.4) along with the encoded amino acid sequence of KRAS B (SEQ ID NO.: 981). The locations of the most common activating missense mutations within the KRAS amino acid sequence are highlighted in grey. Nucleotide sequences targeted by the exemplar)' interfering RNAs aiR As#l-4 within SEQ ID NO. : 980 are in bold and underlined.
  • FIG. 2 shows an exemplary embodiment of aiKRAS silencing of KRAS and PD-L1 expression in KRAS 1 ⁇ cells and ' KRAS r cells
  • FIG. 2A and FIG, 2B show an exemplary embodiment of MDA-MB-231 cells treated with aiKRAS# l, aiKRAS#2, aiKRAS#3 and aiKRAS#4 (at InM and O. lnM).
  • PD-L1 expression and KRAS silencing were confirmed by Western Blot (FIG. 2A) and quantitative real-time PGR (FIG. 2B). Data with mean and error bars represent mean of standard error (SEM).
  • FIG. 2C shows an exemplary embodiment of KRAS silencing of PD-L1 expression in MDA-MB-231 cells at 0, 8, 24, 32 and 48 hours post- transfection.
  • FIG. 2D shows an exemplary embodiment of aiKRAS silencing of KRAS and PD-L1 expression in KRAS mutant cells (H 58, H460 and H2009) and KRAS wild-type cells (RKO, TCCSIJP) transfected with aiKRAS# l (at InM and O. lnM).
  • the expression of KRAS and PD-L1 was confirmed by Western Blot and quantitative real-time qPCR (data not. shown).
  • FIG. 2E shows an exemplary embodiment of the down-regulation of PD-L1 expression resulting from aiRNA mediated silencing of KRAS gene expression in various KRAS MT cell lines.
  • FIG. 3 shows an exemplary embodiment of aiKRAS silencing of ERK phosphorylation in KRAS mutant (KRAS 1 * 1 ) cells.
  • KR AS MT MDA-MB-231 cells and KRAS WT RKO cells were transfected with aiKRAS#l at InM for 48 hours.
  • FIG, 3A shows an exemplary embodiment of KRAS MT MDA-MB-231 and KRAS 1 RKO cell lysates applied to a phosphokinase array (R&D systems, USA).
  • FIG. 3B shows an exemplary embodiment of phosphorylated ERK1/2 being inhibited only in KRAS*" MDA-MB-231.
  • FIG. 4 shows an exemplary embodiment of the effect of aiKRAS silencing on RAS MEK1 2 ERK1 2 signaling pathway in KRAS MT and K AS ⁇ cells.
  • KRAS*" MDA- MB-231 cells and KRAS ' * 1' RKO ceils were treated with aiKRAS# 1 at InM. After a 48h transfection period, the eel! lysate was applied to a Western Blot, to confirm the amount of total or phosphorylaled MEK and ERK.
  • FIG. 4A shows an exemplary embodiment of KRAS silencing inhibiting MEK/ERK pathway in KRAS MT MDA-MB-231 cells but not in KRAS WT RKO cells.
  • FIG. 4A shows an exemplary embodiment of KRAS silencing inhibiting MEK/ERK pathway in KRAS MT MDA-MB-231 cells but not in KRAS WT RKO cells.
  • FIG. 4B shows an exemplary embodiment of inhibition of PD-L 1 gens expression and ERK1/2 phosphorylation in MDA-MB-231 cells treated with the MEK inhibitor, U0126.
  • PD-L1 expression and total and phosphorylated ERK were analyzed by Western Blot.
  • FIGs. 4C and 4D show an exemplary embodiment of the failure of KRAS silencing to inhibit STATS signaling and nuclear localization of RELA RELB in KRAS** 1" MDA-MB-231 cells treated with aiKRAS# l at lnM. After a 48h transfection period, the amount of nuclear localization of NF-KB (RELA and RELB) (FIG. 4C) or the amount of whole and phosphorylated STATS (FIG. 4D) was determined by Western Blot.
  • RELA and RELB nuclear localization of NF-KB
  • FIG. 4D the amount of whole and phosphorylated STATS
  • FIG. 5 shows an exemplary embodiment of KRAS silencing inhibiting the phosphorylation, the accumulation of ERA- 1 protein as well as the transcriptional activity of FRA- 1 gene.
  • KRAS mutant MDA-MB-231, H358, and H460 cells were treated with asKRAS# l at lnM for 48h.
  • FIG. 5A shows an exemplaiy embodiment of a Western blot of the cell lysate and the detection of total or phosphorylated FRA-1.
  • FIG. SB shows an exemplary embodiment of KRAS mutant MDA-MB-231 cells and KRAS wild type RKO cells treated with either control aiRNA or aiKRAS 1.
  • FIG. SB shows an exemplary embodiment of an analysis of the complexes formed in the RAS mutant or KRAS wild type cells by ELISA assay in the presence of FRA-l-specific antibodies.
  • FIG, 5C shows an exemplary embodiment of a ChlP-qPCR analysis of the PD-L1 enhancer. ChlP was conducted in aiControl or aiKRAS treated KRAS mutant MDA-MB-231 cells and KRAS wild type RKO cells.
  • the predicted PD- LI enhancer sequence was enriched in immunoprecipitated chromatin using an anti-FRA-1 antibody and an anti-cJUN antibody in KRAS mutant cells (Fig. SC Left) but not in KRAS wild type cells (Fig.SD Right), or in chromatin incubated with the negative control IP (normal rabbit IgG). Data with mean and error bars represent mean of standard error (SEM).
  • FIG. 6 shows an exemplary embodiment of the effects of FRA-1 on PD-L1 expression in KRAS mutant cells.
  • KRAS mutant MDA-MB-231 and H460 cells were treated with control aiRNA, aiKRAS, and two different FRA-1 aiRNAs (aiFra-1 # 1, and aiFra-1 #2).
  • PD-L1 and FRA- 1 expression were confirmed using Western Blot (Fig. 6A) and qPCR (Fig. 6B). Data with mean and error bars represent mean of standard error (SEM).
  • FIG. 7 shows an exemplary embodiment of cytotoxic T cell activity against KRAS mutant cancer cells after aiKRAS silencing of PD-L1 expression
  • FIG. 7 shows an exemplary embodiment of cytotoxic T cell activity against KRAS mutant cancer cells after aiKRAS silencing of PD-L1 expression
  • CTL 7A shows an exemplary embodiment of cytotoxic T lymphocyte (CTL) activity against KRAS mntant MDA-MB-23.1 cells transfectecl with. aiKRAS.
  • CMV-specific CTLs were expanded to culture human PBMC with HLA-A*02:01 CMV pp65 peptide.
  • aiRNA-transfected Luc-MDA-MB-231 cells were incubated with or without CMV peptide.
  • Peptide loaded or non-loaded MDA-MB-231 cells were plated into 96-well plates (2000 cells/well).
  • CMV-specific CD 8+ T cells were subsequently added to 96-well plates with Effector : Target (E/T) ratio of 50: 1 and incubated for 24 hours.
  • Live Luc ⁇ MDA-MB-231 cells were measured for intracellular luciferase activity with D-Luciferin K.+ salt. The percent, lysis was then calculated as: (Luminescence of CMV peptide pulsed Luc-MDA-MB-23.1/ Luminescence of CMV peptide un ⁇ pulsed Luc-MDA-MB- 231) x 100.
  • Anti-PD-LI antibody (lOpg/mL) was used as a positive control, Data with mean and error bars represent mean of standard error (SEM). Data were subjected to one- way ANOVA with Dimnett's multiple comparison of means test. Statistical significance is displayed as p-value ** pO.01 and *** pO.001.
  • FIG. 7B shows an exemplary embodiment of PD-L1 cell surface expression on MDA-MB-231 cells after aiKRAS and aiPD-Ll transfection.
  • An exemplary embodiment of the calculated geometric mean of fluorescence intensity (MFI) measured by flo cytometry is depicted at the bottom.
  • FIG. 8A shows an exemplary embodiment of some of the canonical KRAS signaling pathways.
  • growth factor binding to cell-surface receptors results in activated receptor complexes, which contain adaptors such as SHC (SH2-containing protein), GRB2 (growth-factor-receptor bound protein 2) and GAB (GRB2-associated binding) proteins. These proteins can recruit SHP2 and SOS1, a guanine nucleotide exchange f ctors (RAS GEF) protein that can increase RAS-guanosine triphosphate (RAS-GTP) levels by catalyzing nucleotide exchange on RAS.
  • SHC SH2-containing protein
  • GRB2 growth-factor-receptor bound protein 2
  • GAB GAB
  • GTPase activating protein (GAP) neurofibromin (NF1) can bind to RAS-GTP and accelerate the conversion of RAS-GTP to inactive RAS- GDP (guanosine diphosphate), which can terminate signaling.
  • GAP GTPase activating protein
  • NF1 GTPase activating protein
  • MEKV-extraeel ar signal-regulated kinase (ERK) cascade can determine key cellular processes including cell proliferation.
  • RAS can also activate the phosphatidylinositol 3-kinase (PI3K) - ⁇ 3-phosphoinositide-dependent protein kinase 1 (PDK1)- AKT pathway that can determine cellular survival.
  • PI3K phosphatidylinositol 3-kinase
  • PDK1- AKT pathway that can determine cellular survival.
  • RALGDS, RALGDS-like gene (RGL), RGL2 and TIAM1 can be exchange factors of RAL and RAC, respectively.
  • RAL phospholipase D
  • RAC can regulate actin dynamics and, therefore, the cytoskelelon.
  • RAS can also bind and activate the enzyme phospholipase Cepsilon (PLCep ' silon), the hydrolytic products of which can regulate calcium signaling and the protein kinase C (PKC) family.
  • FIG, 8B shows a proposed signaling mechanism along the KRAS/ME ERK Fra- 1 PD-L 1 axis, hi KRAS mutant cancer cells, mutant KRAS can activate ME ERK kinase.
  • Stabilized phosphorylated FRA-1 can bind to the PD-L1 enhancer region where it can activate and maintain FD-L1 gene expression in tumor cells.
  • the phrase "at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combi nations of elements in the list of elements.
  • This definition also allows mat elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase "at least one" refers, whether related or unrelated to those elements specifically identified.
  • "at least one of A and B" can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with mo A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
  • the term “about” when used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below those numerical values.
  • the term “about” is used herein to modify a numerical value above and below the stated value by a vari nce of 20%, 10%, 5%, or 1%, in certain embodiments, the term “about” is used to modify a numerical value above and below the stated value by a variance of 1.0%.
  • the term “about” is used to modify a numerical value above and below the stated value by a variance of 5%.
  • the 'term “about” is used to modify a numerical value above and below the stated value by a variance of 1%,
  • a "polynucleotide” refers to a polymeric chain containing two or more nucleotides. "Polynucleotides” includes primers, oligonucleotides, nucleic acid strands, etc. A polynucleotide ma contain standard or non-standard nucleotides. Typically, a polynucleotide contains a 5' phosphate at one terminus ("5' terminus”) and a 3' hydroxy! group at the other terminus ("3* terminus) of the chain.
  • the most 5' nucleotide of a polynucleotide may be referred to herein as the "5 -term.inal nucleotide” of the polynucleotide.
  • the most 3' nucleotide of a polynucleotide may be referred to herein as the "3 '-terminal nucleotide” of the polynucleotide,
  • subject generally refers to an organism to which a compound or pharmaceutical composition described herein can be administered.
  • a subject can be an animal or animal cell, including a mammal or mammalian cell (e.g., a human or human cell).
  • the term also refers to an organism, which inchjdes a cell or a donor or recipient of such cell.
  • the term "subject” refers to any animal (e.g., a mammal), including, but not limited to, humans, mammals and non-mammals, such as non-human primates, mice, rabbits, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, fish, nematode, and insects, which is to be the recipient of compound or pharmaceutical composition described herein.
  • a mammal e.g., a mammal
  • mammals and non-mammals such as non-human primates, mice, rabbits, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, fish, nematode, and insects, which is to be the recipient of compound or pharmaceutical composition described herein.
  • the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.
  • administer refers to any method of introducing to a subject a compound or pharmaceutical composition described herein and can include, for example, introducing a compound systemically, locally, or in situ to the subject.
  • a compound of the present disclosure produced in a subject from a composition is encompassed by these terms.
  • systemic or “systemically,” they generally refer to in vivo systemic absorption or accumulation of the compound or composition in the blood stream and its distribution throughout the entire body.
  • the terms "administer,” “administering.” or “administration” can refer to, for example, delivering one or more recombinant vectors to a tumor cell, wherein the vector expresses an RNA interfering agent as defined herein.
  • the tumor cell is in a subject.
  • combination treatment mean the administration of at least two different agents (e.g., at least one compound chosen from modulators of KRAS signaling and/or at least one compound chosen from therapeutic agents, and, optionally, one or more additional agents) to treat a disorder, condition, or symptom, e.g., a cancer condition.
  • agents e.g., at least one compound chosen from modulators of KRAS signaling and/or at least one compound chosen from therapeutic agents, and, optionally, one or more additional agents
  • combination treatment may involve the administration of one agent before, during, and/ or after the administration of a second agent.
  • the first agent and the second agent can be administered concurrently, separately, or sequentially in separate pharmaceutical compositions.
  • a treatment combination comprises a therapeutically effective amount of at least one compound chosen from modulators of KRAS signaling and a therapeutically effective amount of at least, one compound chosen from therapeutic agents, e.g. immune checkpoint inhibitors.
  • the immune checkpoint inhibitor can be. for example, an inhibitor of PD-L1.
  • the at least one compound chosen from modulators of KRAS signaling and at least one compound chosen from therapeutic agents can have different mechanisms of action.
  • a combination treatment improves the prophylactic or therapeutic effect of the at least one compound chosen from modulators of KRAS signaling and the at least one compound chosen from therapeutic agents by functioning together to have an additive, synergistic, or enhanced effect.
  • a combination treatment of the present disclosure reduces adverse side effects associated with the at least one compound chosen from modulators of KRAS signaling and the at least one compound chosen from therapeutic agents.
  • the administration of the at least one compound chosen from modulators of KRAS signaling and the at least, one compound chosen from therapeutic agents may be separated in time by up to several weeks, but more commonly within 48 hours, and most commonly within 24 hours.
  • a "therapeutic agent” that may administered with a modulator of KRAS signaling can be an anticancer therapeutic agent, i.e. an agent that may be administered in vivo to treat cancer.
  • the anticancer therapeutic agent can be a small molecule, a peptide, a modified peptide, a peptidomimetic, an antibody, an antibody fragment, a recombinant antibody, a recombinant antigen-binding protein, an aptamer, a nucleic acid or RNA interfering agent,
  • the anticancer therapeutic agent can be, for example, a chemotherapeutic agent, i.e. a chemical compound useful in the treatment of cancer.
  • chemotherapeutic agents include, but are not limited to, alkylating agents, antimetabolites, kinase inhibitors, mitotic inhibitors, spindle poison plant alkaloids, cytotoxic /antitumor antibiotics, topisomerase inhibitors, photosensitizers, anti-estrogens and selective estrogen receptor modulators (SERMs), anti-progesterones, estrogen receptor down-regulators ERDs), estrogen receptor antagonists, luteinizing hormone-releasing hormone agonists, anti- androgens, aron atase inliibitors, EGFR inliibitors, angiogenesis inhibitors, VEGF inhibitors, and inhibitors of the translation and/or transcription of genes implicated in abnormal cell proliferation or tumor growth.
  • Chemotherapeutic agents useful in the treatment methods disclosed herein include cytostatic and/or
  • the therapeutic agent can be, for example, a biotherapeutic agent, such as an antibody or recombinant antigen-binding protein, in certain embodiments, the antibody or recombinant antigen-bind protest! can block ligand / receptor signaling its any biological pathway that supports tumor maintenance and/or growth or suppresses the antitumor immune response.
  • the therapeutic agent can be, for example, a targeted therapeutic agent, such as a small molecule drug, hi certain embodiments, the therapeutic agent can include radiation and/or surgery.
  • the therapeutic agent can be, for example, a small molecule such as a small molecule kinase inhibitor (SMKI), SM Is that can be combined with a modulator of KRAS signaling are disclosed, for example, in Wu et al Drug Discovery Today (2016) volume 21, issue 1, pp. 5-10, which is incorporated herein by reference.
  • SMKI small molecule kinase inhibitor
  • Exemplary SMKIs are depicted in TABLE 1 below (dates indicate year when FDA approved).
  • effective amount and “therapeutically effective amount” refer to the amount of a compound or pharmaceutical composition described herein that is capable of invoking, for example, one or more of the following effects: (1) inhibition, to some extent, of cancer or tumor growth, including a decrease or cessation in the progression of cancer; (2) reduction in the number of cancer or tumor cells; (3) reduction in tumor size; (4) inhibition, e.
  • a decrease or a cessation, of cancer or tumor cell infiltration into peripheral organs e.g., a decrease or a cessation, of cancer or tumor cell infiltration into peripheral organs
  • inliibition e.g., a decrease or a cessation, of metastasis
  • enhancement of anti-tumor immune response which may, but is not required to, result in the regression or rejection of the tumor, or (7) relief, to some extent, of one or more symptoms associated with the cancer or tumor.
  • the therapeutically effective amount can vary depending upon the intended application (in vitro or in vivo), the subject and disease condition being treated, e.g., the sex, weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which cars readily be determined by one of ordinary skill m the art, e.g., a board-certified oncologist.
  • a 'therapeutically effective amount is an amount of a compound where any toxic or detrimental effects resulting from the administration of the compound are outweighed by the therapeutically beneficial effects.
  • progress refers to at least one of the following: (1) a response of progressive disease to prior therapy (e.g., chemotherapy and/or immune checkpoint therapy); (2) the appearance of one or more new lesions after treatment with prior therapy (e.g.. chemotherapy and/or immune checkpoint therapy); and (3) at least a 5% (e.g., 10%, 20%) increase in the sum of diameters of target lesions, taking as a reference the smallest sum on study.
  • prior therapy e.g., chemotherapy and/or immune checkpoint therapy
  • a 5% e.g. 10%, 20%
  • the term "sensitize” means to alter cancer cells or tumor cells in a way that allows for more effective treatment of the associated cancer with a cancer therapy.
  • normal cells are not affected to an extent that causes the normal cells to be unduly injured by the cancer therapy.
  • an increased sensitivity or a reduced sensitivity to a therapeutic treatment can be measured according to a known method in the art for the particular treatment and methods described herein below, including, but not limited to, cell proliferative assays (Tanigawa et ah Cancer Res 1982; 42: 2159-2164) or cell death assays (Weisenthal et al Cancer Res 1 84; 94: 161.
  • the sensitivity or resistance may also be measured in animals by measuring the tumor size reduction over a period of time, for example, 6 months for humans and 4-6 weeks for mice.
  • a composition or a method sensitizes cancer cells or tumor cells to a therapeutic treatment if the increase in treatment sensitivity or the reduction in resistance is about 25% or more, for example, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90% or more, compared to treatment sensitivity or resistance in the absence of such composition or method.
  • the increase in treatment sensitivity or the reduction in resistance is about 2-fold, about 3-fold, about 4-fold, about 5-fold, about 10-fold, about 15-fo!d, about 20-fold or more compared to treatment sensitivity or resistance in the absence of such composition or method.
  • the determination of sensitivity or resistance to a therapeutic treatment is routine in the art and within the skill of an ordinarily skilled clinician. It is to be understood that any method described herein for enhancing the efficacy of a cancer therapy can be applied to methods for sensitizing hyperproliferative or otherwise cancerous cells (e.g., resistant cells) to the cancer therapy.
  • the terra "synergy,” “synergistic,” “synergisticaily,” or “enhanced” as used herein refers to an effect of interaction or combination of two or more components to produce a combined effect greater than the sum of their separate effects (or “additive effects”).
  • a synergistic effect may be attained when the compounds are: (.1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. in separate tablets, pills or capsules, or by different injections in separate syringes.
  • a synergistic anticancer effect denotes an anticancer effect which is greater than the predicted purely additive effects of the individual compounds of the combination administered separately.
  • Terms such as “treating” or “treatment” or “to treat” as used herein refer to (1) therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder . or/and (2) prophylactic or preventative measures that prevent or slow the development and/or progression of a targeted pathologic condition or disorder,
  • those in need of treatment include those already with the disorder; those prone to have the disorder; and those in whom the disorder is to be prevented
  • a subject is successfully "treated” according to the methods of the present disclosure if the patient shows one or more of the following: a reduction in the number of or complete absence of cancer cells; a reduction in tumor size; inhibition of or an absence of cancer cell infiltration into peripheral organs including the spread of cancer into soft tissue and bone; inhibition of or an absence of tumor metastasis: inhibition or an absence of tumor growth; relief of one or more symptoms associated with a specific cancer; reduced morbidity and mortality; and improvement in quality of life.
  • a “modulator” refers to a compound or combination of compounds that is capable of modulating KRAS signaling activity, including but not limited to, oncogenic KRAS signaling or otherwise aberrant KRA8 signaling activity,
  • a “modulator” can refer to a compound or combination of compounds that is capable of modulating the expression a target gene required for KRAS signaling.
  • a “modulator” can refer to a compound or combination of compounds thai are capable of modulating the expression of KRAS.
  • a “modulator” can refer to a compound or combination of compounds that, are capable of modulating tine expression of one or more of KRAS, RAF, MEK, ERK and FRA- 1.
  • modulating and its grammatical equivalents refer to either increasing or decreasing (e.g., silencing), in other words, either up-regulating or down-regulating KRAS signaling activity, e.g., by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, 99% or 100%, compared to KRAS signaling activity in the absence of a modulator.
  • a “modulator” can refer to an inhibitor of a cellular activity, e.g, an inhibitor of oncogenic KRAS signaling.
  • a “modulator” can refer to an activator of a cellular activity, e.g. RAS GAP induced ' hydrolysis of GTP bound to KRAS.
  • the terms “inhibiting”, “to inhibit” and their grammatical equivalents, when used in the contest of a bioactivity, refer to a down-regulation of the bioactivity, which may reduce or eliminate the targeted function, such as the production of a protein or the phosphorylation of a molecule.
  • the terms refer to a down-regulation of a bioactivity of the organism, which may reduce or eliminate a targeted function, such as the production of a protein or the phosphorylation of a molecule, in particular embodiments, inhibition may refer to a reduction, e.g., of about 10%, of about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or about 100% of the targeted activity.
  • the terms refer to success at preventing the onset of symptoms, alleviating symptoms, or eliminating the disease, condition or disorder.
  • cancer h a subject refers to the presence of cells possessing characteristics typical of cancer-causing ceils, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain morphological features. Often, cancer cells will he in the form of a tumor or mass, hut such cells may exist alone within a subject or may circulate in the blood stream as independent cells, such as leukemic or lymphoma cells.
  • cancer examples include, but are not limited to lung cancer, pancreatic cancer, bone cancer, skin cancer, head or neck cancer, cutaneous or intraocular melanoma, breast cancer, uterine cancer, ovarian cancer, peritoneal cancer, colon cancer, tnicrosatellite instability-high metastatic colorectal cancer, microsatellite stable metastatic colorectal cancer, colorectal cancer with mismatch-repair deficiency, colorectal cancer without mismatch-repair deficiency, small bowel adenocarcinoma, rectal cancer, colorectal adenocarcinoma, cancer of the anal region, stomach cancer, gastric cancer, gastrointestinal cancer, gastric adenocarcinoma, adrenocorticoid carcinoma, genitourinary cancer, gynecologic cancer, uterine cancer, uterine sarcoma, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the vagina
  • urological cancer a general term, includes bladder cancer, prostate cancer, kidney cancer, testicular cancer, and the like
  • hepatobiliary cancer another general term, includes liver cancers (itself a general term that includes hepatocellular carcinoma or cholangiocarcmoma), gallbladder cancer, biliary cancer, or pancreatic cancer. Both urological cancer and hepatobiliary cancer are contemplated by the present disclosure and included in the term "cancer.”
  • solid tumor refers to those conditions, such as cancer, that form an" abnormal tumor mass, such as sarcomas, carcinomas, and lymphomas.
  • solid tumors include, but are not limited to, non-small cell lung cancer (NSCLC), neuroendocrine tumors, th omas, fibrous tumors, metastatic colorectal cancer (mCRC), and the like, in certain embodiments, the solid tumor disease is an adenocarcinoma, squamous cell carcinoma, large cell carcinoma, and the like.
  • the cancer is breast cancer. In certain embodiments, the cancer is colorectal adenocarcinoma. In certain embodiments, the cancer is small bowel adenocarcinoma. In certain embodiments, the cancer is hepatocellular carcinoma. In certain embodiments, the cancer is head and neck cancer. In certain embodiments, the cancer is renal cell carcinoma. In certain embodiments, the cancer is ovarian cancer. In certain embodiments, the cancer is prostate cancer. In certain embodiments, the cancer is lung cancer. In certain embodiments, the cancer is uterine sarcoma. In certain embodiments, the cancer is esophageal cancer. In certain embodiments, the cancer is pancreatic cancer. In certain embodiments, the cancer is a gastric cancer.
  • the cancer is endometrial cancer. In certain embodiments, the cancer is cholangiocarcinoma. In certain embodiments, each of the cancers is unresectable, advanced, refractory, recurrent, or metastatic. In certain embodiments, the cancer is resistant or has acquired resistance to an anticancer therapeutic agent, e.g. treatment with an immune checkpoint inhibitor.
  • the efficacy of a compound or a combination of compounds is tested in a xenograft cancer model in which cells isolated from a solid tumor are injected into a host animal, e.g. an immunocompromised host, to establish solid tumors.
  • the cells isolated from a solid tumor comprise cancer stem cells.
  • the host animal can be a model organism such as nematode, fruit fly, zebrafish; preferably a laboratory mammal such as a mouse (nude mouse, SCID mouse, or NOD/'SCID mouse, Beige /SCED Mouse), rat, rabbit, or primate.
  • the severely immunodeficient NOD-SCID mice ma be chosen as recipients to maximize the participation of injected cells.
  • a KRAS gene (also called C-K-RAS; CFC2; K-RAS2A; K-RAS2B; K-RAS4A; K-RAS4B; KI- RAS; KRASl ; KRAS2; NS; NS3; RASK2) encodes tire human cellular homoiog of a transforming gene isolated from the Kirsten rat sarcoma virus.
  • KRAS is a member of the mammalian RAS gene family that encode a group of closely related 21 kDa GDP/GTP-binding proteins that can act as intracellular signal transducers.
  • KRAS protein refers to a polypeptide having at least about 40%, e.g., about 80%, identity to the amino acid sequence provided at Genbank Accession No. AAB41942 or ABY87538.
  • the KRAS protein can refer to a polypeptide comprising at least 10 contiguous amino acids of the amino acid sequence of SEQ ID NO. : 981.
  • the KRAS protein refers to the KRAS isoform B having the exemplary amino acid sequence of SEQ ID NO. : 981. 1 Si SKSiW S JM3SV»3 ⁇ 4CS&ldi QS.XQ3 ⁇ 4HF ⁇ B STOSFTXEDSX KKQWVXOQftF ⁇ 3 ⁇ 4 ⁇ 3 ⁇ 4» ⁇ . ⁇ 3 ⁇ 4
  • the N-terminal portion (residues 1- 165) of KRAS comprises a highly conserved G domain which is also found in H-RAS and N-RAS isoforms.
  • RAS proteins can diverge substantially at the C- terminal end, which is known as the hypervariabl e region. This region can contain residues that specify post-translational protein modifications that are essential for targeting RAS proteins to the cytosolic leaflet of cellular membranes. All RAS proteins are famesylated at a terminal CAAX motif, in which C is cysteine, A is usually an aliphatic amino acid, and X is any amino acid.
  • KRAS4A is additionally modified by one or two palmitic acids just upstream of the CAAX motif.
  • the addition of the hydrophobic farnesyl moiety is complemented by the hydrophobic palmitates (the so-called "second signal") to firmly anchor KRAS4A to the membrane.
  • KRAS4B the predominant splice variant, contains an alternative second signal that is composed of a polybasic stretch of lysine residues.
  • membrane anchoring is mediated by the electropositive lysines that form ionic bonds to the predominantly electronegative lipid head groups of the inner leaflet of the plasma membrane.
  • KRAS encompasses wild type KRAS 4 A and/or KRAS4B. In certain embodiments, the term “KRAS” encompasses both wild type and modified forms of KRAS 4 A. and/or KRAS4B.
  • modified forms of KRAS include, but are not limited to, KRAS proteins having one or more activating mutations, for example, missense mutations at positions G12, G13 and/or Q61.
  • KRAS encompasses KRAS proteins having one or more alterations in the post-translational modifications of KRAS, including, but not limited to, acetylation, methylation, lipidation, palmitoylation, prenyl ati on, and S- nitrosylation.
  • KRAS Tethered to the inner leaflet of the plasma membrane, KRAS can act as a binary molecular switch at the apex of a signaling hub where it can control the transmission of signals from cell surface receptors to intracellular effectors by cycling between a GDP -bound inactive and a transient GTP-bound active slate. In its active GTP-bound form, KRAS can activate downstream effectors that control cellular processes in the cytoplasm (actin organization, endocytosis) or modify the activity of nuclear transcription factors that, regulate gene expression important for cell cycle progression, differentiation, or survival.
  • the KRAS molecular switch can function by responding to upstream signals by activating a class of proteins known as guanine nucleotide exchange factors (RAS GEFs) that can stimulate the dissociation of GDP from the RAS protein.
  • RAS GEFs guanine nucleotide exchange factors
  • SOS I a RAS GEF in the MAPK/ER pathway
  • GRB2 an adaptor protein GRB2 in response to epidermal growth factor receptor (EGFR) activation.
  • EGFR epidermal growth factor receptor
  • GTPase acting proteins like pl2QGAP., that enhance GTP hydrolysis by the otherwise slow intrinsic GTPase activity of RAS proteins.
  • RAS-GTP can preferentially bind to and activate downstream RAS- binding-domain (RBD) or RAS-association (RA)-domain-containing effectors. It can be estimated that there are at least 11 distinct RAS effector families, each of which can activate a distinct protei n signaling cascade. Exemplary downstream effector pathways that respond to the KRAS activation are depicted in FIG. 8A and summarized below.
  • GTP-bound KRAS One exemplary downstream effector of activated GTP-bound KRAS is the RAS-RAF- MAP-MEK-ERK kinase cascade which can be an essential, shared element of mitogenic signaling involving tyrosine kinase receptors that leads to a wide range of cellular responses, including growth, differentiation, inflammation, and apoptosis
  • GTP-bound R AS can recruit RAF serine/threonine kinases (A-RAF, B-RAF and C-RAF- 1)
  • A-RAF, B-RAF and C-RAF- 1 RAF serine/threonine kinases
  • the interaction of activated GTP-bound KRAS with RAF can initiate the RAF ⁇ MEK ⁇ ERK kinase cascade.
  • Activated RAF a MAPK (Mitogen-activated protein) kinase-kinase
  • MAPK Mitogen-activated protein
  • EK1 and MEK2 also known as MAP2K1 and MAP2K2
  • ERK1 and 2 extracellular signal-regulated kinase
  • Activated ERK1/2 can then phosphorylate many substrates, including kinases that are important for control of translation (e.g., p90RSK) and transcription factors thai control genes involved in cell cycling (e.g., ELK1, FOS, MYC, FRA-1)
  • PBKs are heterodimeric lipid kinases composed of a regulatory subunit (p85) and a catalytic subunit (pi 10).
  • GTP-bound RAS bound to the pi 10 (o, ⁇ , ⁇ , and ⁇ ) catalytic subunits of class I PBK can trigger the synthesis of the secondary messenger, phophatidylinositol-3,4 s 5-triphosphate (PIP3), PIPS can then be free to engage the pleckstrin homology (PH) domain of AKT/PKB (Protein kinase B), thereby stimulating its Ser/Thr kinase activity and the phosphorylation of a host of other proteins involved in cell growth, cell cycle entry, and cell survival.
  • AKT-raediated phosphorylation can inhibit some proteins that promote programmed cell death (BAD, FoxO), and stimulates others (MDM2) that, promote cell survival.
  • RAS effectors include RAIN, an endomembrane receptor for RAS, NOREL a pro-apoptotic tumor suppressor, and AF-6, a mediator of membrane-cytoskeleton interactions.
  • RAS can also engage in cross- talk with other GTPase signaling pathways involved in regulating acttn reorganization and/ or endocytic trafficking. This can occur through interactions of RAS with RAL guanine nucleotide dissociation stimulator for the RA8-!ike (RAL) small GlPases, RALA and RALE, a GEF that facilitates GDP/GTP exchange with RAS (RALGDS).
  • RALGDS a GEF that facilitates GDP/GTP exchange with RAS
  • RAS can also interact with GEFS like RINl, for RAB5, and TIAM.1 (tumor invasion and metastasis inducing protein 1), for RAC.
  • GEFS like RINl
  • TIAM.1 tumor invasion and metastasis inducing protein 1
  • accumulating evidence indicates signaling specificity may also be dictated by differential localization of KRAS isoforms in discrete plasma membrane microdomains or distinct intracellular membrane compartments (e.g., endosomes, Golgi), where the activated GTPase may encounter a unique sets of effectors,
  • KRAS can be essential for mammalian embryonic development. KRAS -deficient mice can die of anemia and defective fetal liver erythropoiesis after only about 12-14 days of gestation. Germline mutations that affect components of the RAS-RAF— MEK-ERK pathway can cause several developmental disorders, including Noonan Syndrome (NS3), Costello Syndrome and Cardio-Facio-Cutaneous (CFC2) syndrome. The developmental disorders associated with RAS pathway mutations may share phenotypic features that include facial abnormalities, heart defects, impaired growth and development, and. in some instances, a predisposition to specific cancers.
  • NS3 Noonan Syndrome
  • CFC2 Cardio-Facio-Cutaneous
  • KRAS is also a proto-oncogene
  • a proto-oncogene can become oncogenic by increased KRAS expression or the acquisition of an "activating" mutation, i.e. a mutation that leads to constitutive aberrant activation of KRAS signaling. Indeed, activating somatic KRAS mutations have been detected in—30% of all human cancers. Amongst those cancers.
  • KRAS cart be found in a predominantly mutated form in pancreatic ductal adeno-carcinoma (71 %), colorectal cancer (35%), non-small cell lung adenocarcinoma (19%), and endometrial cancer (17%) (see TABLE 2 below; collated from the Catalogue of Somatic Mutations in Cancer (COSMIC) database).
  • Activating KRAS gene point mutations can also be present in other cancers, including but not limited to, biliary tract malignancies, endometrial cancer, cervical cancer, bladder cancer, liver cancer, myeloid lexikemia and breast cancer.
  • Mutant KRAS refers to somatic or germlme KRAS4 A and/or K AS4B mutations including, but not limited to, point mutations, nonsense substitutions, missense substitutions, synonymous substitutions, in frame insertions, frameshift insertions and/or deletions, KRAS missense gain-of -function activating mutations can be found predominantly at one of three mutational hotspots: G12 (89%), G13 (9%), and Q61 (1%). In certain embodiments, these mutations disrupt intrinsic as well as GAP-mediated GTP hydrolysis. In certain embodiments, the disruption results in an accumulation of constitutive! y active GTP- bound RAS in cancer cells.
  • a mutant KRAS can refer to aberrant post-translational modification of KRAS.
  • the aberrant post-translational modification of KRAS includes, but not limited to. phosphorylation, glycosylation, ubiquitination, nitrosylation, methylation, acetylation, lipidation (C-terminal glycosyl phosphatidylinositol (GPI) anchor, N-tercninal myriatoylation, S-myristoylation, S-prenylation) and/or proteolysis.
  • a mutant KRAS can refer to aberrant splicing of KRAS mRNAs.
  • KRAS4B mutations associated with cancer include, without limitation, KRAS G52D , KRAS GI2V , KRAS GUD , KRAS G12C , KRAS Q61R , KRAS3 ⁇ 4 S1L , KRAS3 ⁇ 4 6IK , KRAS Gi 2R , and KRAS G12C .
  • KRAS gene comprising a different KRAS mutation than one of those above and/or combinations of the above and/or other KRAS mutations that lead to constitutive activation of KRAS signaling, is also an oncogenic KRAS encompassed by the present disclosure,
  • a comprehensive list of KRAS mutations present in human cancer is available online from UniProt Consortium, EMBL.
  • KRAS signaling can refer to wild ty e KRAS4A and/or KRAS4B GTPase activity, GTP/GDP binding activity or any signaling activity induced by ⁇ GTP-bound KRAS4A and or KRAS4B, including, but not limited to, the RAS-RAF- AP- MEK-ER , the RAS-PDKs-AKT and RAS-RalGDS signal transduction pathways as summarized in. part above (e.g., see FIG, 8 A).
  • KRAS signaling encompasses any form of aberrant KRAS signaling.
  • aberrant KRAS signaling can occur as a result of sig aling by an oncogenic KRAS protein having one or more activating mutations, including, but not limited to, missense mutations at positions G12, GI3 and/or Q61.
  • the aberrant KRAS signaling refers to the activity of a hyperactive wild type GTP bound KRAS as a result of changes in GDP--GTP regulation, loss of GAPs or persistent receptor tyrosine kinase-mediated activation of GEFs.
  • aberrant KRAS signaling can be caused by the aberrant, activation of effector molecules downstream of KR AS, including, but not limited to, A-RAF. B-RAF, C-RAF, MEK, ERK and/or F.RA-L
  • aberrant KRAS signaling e.g. oncogenic KRAS signaling, can occur in the presence of wild type KRAS.
  • KRAS signaling comprises hyperactive KRAS signaling initiated as a result of the inactivation of a tumor suppressor that, when inactivated, provides an alternative mechanism of activating RAS
  • the hyperactive KRAS signaling results from the inactivation of a tumor suppressor such as a RAS GAP
  • RAS GAPs include, but are not limited to, RASAl, RASA2, RAS A3, RASA4, RASALl, NFl , DAB2IP, RASAL2, RASAL3, SynGAPl, IQGAP1, IQGAP2 and IQGAP3.
  • mutant or otherwise modified KRAS has been implicated in developmental disorders and virtually all aspects of the malignant phenotype of the cancer cell, including cellular proliferation, transformation, invasion and metastasis.
  • oncogenic KRAS in the etiology of human cancers, efforts to develop small molecule drugs targeting, for example, oncogenic KRAS over the past three decades have been largely unsuccessful.
  • MAP kinase-kinase (MEK) inhibitors and phosphatidylinositol 3-kinase (PI3K) inhibitors have not yet shown significant clinical activity in RAS associated cancers, for reasons relating to feedback loops and poor therapeutic windows as well as lack of specificity. This failure has led some to dismiss KRAS as an "undruggable" target. Even strategies employing siR s to target mutant KRAS remain challenging primarily because of off target silencing of genes unrelated to KRAS or the induction of a robust interferon response.
  • the present disclosure reports on an approach for specifically inhibiting aberrant KRAS signaling in tumor cells without the known caveats associated with RNA interference.
  • oncogenic KRAS signaling in KRAS associated cancer cell lines can induce aberrant RAF MEK ERK/F A-1 signaling that can stimulate the constitutive high level expression of the immune checkpoint, PD-L1.
  • the interaction of PD-L1 expressed on the surface of tumor cells with PD-1 receptor on T cells ca trigger the activation of the PD-Ll/PD- 1 immune checkpoint pathway in T cells which can lead to the suppression of tumor cell-specific T eel!
  • the targeted inhibition of K AS signaling e.g., by RAS-specific asymmetric interfering RNAs, can down-regulate PD-L1 gene expression and restore the sensitivity of cancer ceils expressing an oncogenic 5 KRAS to killing by antigen-specific cytotoxic T cells (see Example 4).
  • tire inhibition of oncogenic KRAS signaling in cancer cells can inhibit MEK/ERK-dependenl phosphorylation
  • the inhibition of oncogenic KRAS signaling in cancer cells can inhibit the accumulation of FRA.-1 protein
  • FRA-1 protein is a transcription factor required for the activation of the AP-1 responsive enhancer 10 within the first intron of the PD-L1 gene, Inhibition of KRAS signaling may therefore provide a novel approach to sensitizing cancer cells resistant to immunotherapies, such as immune checkpoint therapies, as well as improving the efficacy of known anticancer therapeutics.
  • the modulator of ICR AS signaling may comprise, for example, an inhibitor that reduces or prevents KRAS-mediated cell signaling in tumor cells, in certain I S embodiments, the tumor cell expresses an oncogenic KRAS, in certain embodiments, the inhibitor can directly target both wild type and/or oncogenic KR AS by inhibiting the expression of KRAS in tumor cells.
  • the inhibitor of KRAS can be, for example, an RNA interfering agent.
  • RNA interfering agent is defined as any agent that can inhibit the 0 expression of a target gene by RNA. interference (RNAi).
  • RNA interference is an evoiutionally conserved process whereby the expression or introduction of an RNA comprising a sequence that is identical to or highly similar to a target gene sequence can result in the sequence-specific degradation or specific post-transcriptional 5 gene silencing (PTGS) of messenger RNA (mRNA) transcribed from that targeted gene, thereby iiimbiting target gene expression, in nature, RNAi is initiated by the dsRNA-specific endonuclease, Dicer, a member of RNase III rihonuclease family.
  • PTGS post-transcriptional 5 gene silencing
  • Dicer cleaves long, double- stranded RNA (dsRNA), pre-microRN A (miRNA), and short hairpin RNA (shRNA) into short double-stranded RNA fragments called small interfering RNAs (siRNA) of about 20-25 0 nucleotides in length, usually with a two-base overhang on the 3' end.
  • Dicer catalyzes the first step in the RNA interference pathway and initiates formation of the RNA-induced silencing complex (RISC), whose catalytic component, argonauts, is an endonuclease capable of degrading messenger RNA (mRNA) whose sequence is complementary to that of the siRNA guide strand.
  • RISC RNA-induced silencing complex
  • RNAi can be initiated by introducing nucleic acid molecules, e.g., synthetic siRNAs or RNA interfering agents, having a guide RNA thai targets a specific expressed gene sequence.
  • RNA interfering agents include, but are not limited to, small non-coding RNAs such as antisense oligonucleotides, shRNAs (e.g. as disclosed in U.S. Patent No. 7,750, 144, which is incorporated by reference herein in its entirety for any purpose), siRNAs (e.g. as disclosed in U. S.
  • Patent No, 7,056,704 and 9,260,470 which are incorporated by reference herein in their entireties for any purpose
  • a microRNA or a mature microRNA molecule or a pre-microRNA molecule or a primary microRNA molecule, or a variant thereof e.g. as disclosed in U. S. Patent No. 8,609,831, which, is incorporated by reference herein in its entirety for any purpose
  • gapraers e.g. as disclosed in U. S. Patent No. 6,107,094, which is incorporated by reference herein in its entirety for any purpose
  • IncRNA e.g. as disclosed in the International Publication No.
  • WO2012018881 which is incoiporated by reference herein in its entirety for any purpose
  • a piRNA (piwiR A) molecule e.g. as disclosed in the International Publication No. WO2008109142, which is incorporated by reference herein in its entirety for any purpose
  • a triplex oligonucleotide e.g. as disclosed in U.S. Patent. No. 5,693,773, which is incorporated by reference herein in its entirety for any purpose
  • ribozym.es e.g. as disclosed in U.S. Patent No. 5,225,347, which is incorporated by reference herein in its entirety for any purpose).
  • RNA interfering agents Exemplary chemical modifications of RNA interfering agents are disclosed in Dar et al siRNAmod: A database of experimentally validated chemically modified siRNAs. Sci. Rep. (2016) 6, 20031.
  • compositions comprising a class of short double stranded RNA.
  • interfering agents called asymmetrical interfering RNAs (aiRNA)
  • aiRNA asymmetrical interfering RNAs
  • PCX Publications WO 2009/029688 and WO 2009/029690 the contents of which are hereby incorporated by reference in their entireties for any purpose.
  • this class of RNAi- inducers is characterized in the length asymmetry of the two RNA strands.
  • aiRNA can have RNA duplex structure of much shorter length than the other siRNA, which should reduce the cost of synthesis and abrogate/reduce the length-dependent triggering of nonspecific interferon- like responses.
  • the asymmetry of the aiRNA structure abrogates and/or otherwise reduces the sense-strand ' mediated off-target effects. aiRNA is therefore, in certain embodiments, more efficacious, more potent, with a more rapid-onset, and more durable ai inducing gene silencing than any of the other RNA interfering agents.
  • aiRNAs disclosed herein each comprises a first strand with a length from 18-23 nucleotides (nt) and a second strand with & length from 12-17 nucleotides.
  • the second strand is substantially complementary to the first strand.
  • the second strand forms a double-stranded region with the first strand, in certain embodiments, the first strand has a 3 -overhang from 1-9 nucleotides.
  • the first strand has a 5 -overhang from 0-8 nucleotides.
  • the aiRNA is capable of effecting at silencing KRAS signaling in a eukaryotic cell.
  • the first strand is 18, 19, 20, 22, or 23 nucleotides long.
  • the second strand is 12, 13, 14, 15, 16, or 17 nucleotides long. In certain embodiments, the 3 -overhang is greater than 0 nucleotides in length. In certain embodiments, the first strand comprises a sequence being substantially complementary to a target KRAS mRNA sequence. In certain embodiments, the first strand comprises a sequence being at least 70 percent complementary to a target mRNA sequence.
  • the first strand is at least 1 nt longer than the second strand.
  • the firs strand is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nt longer than the second strand.
  • the first strand is 20-100 nt longer than the second strand.
  • the first, strand is 2-12 rit longer than the second strand.
  • the first strand is 3-10 nt longer than the second strand.
  • the first strand, or the long strand has a length of 5-100 nt, or preferably 10-30 or 12-30 nt, or more preferably 15-28 nt. In one embodiment, the first strand is 21 nucleotides in length. In some embodiments, the second strand, or the short strand, has a length of 3-30 nt, or preferably 3-29 nt or 10-26 nt, or more preferably 12-26 nt. In some embodiments, the second strand has a length of .15 nucleotides.
  • the double-stranded region has a length of 3-98 base pairs (bp).
  • the double-stranded region has a length of 5-28 bp. In an even further embodiment, the double-stranded region has a length of 10-19 bp, The length of the double- stranded region can be 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 bp.
  • aiRNAs disclosed herein comprise a duplex RNA comprising a first strand, wherein the 5 '-terminal and 3 '-terminal nucleotides of the first strand are from 16 to 21 nucleotides apart, and a second strand, wherein the 5 '-terminal and 3 '-terminal nucleotides of the second strand are from 10 to 17 nucleotides apart, wherein the 5 '-terminal nucleotide of the second strand is complementary to a nucleotide of the first strand other than its 3 '-terminal nucleotide, wherein the 3 '-terminal nucleotide of the second strand is complementary to a nucleotide of the first, strand,
  • the penultimate nucleotide from the 3 '-terminal nucleotide of the first strand is not dT.
  • the duplex RNA is more effective at silencing an expressed nucleotide sequence of a target gene than a corresponding 21-mer siRNA duplex targeting the same expressed nucleotide sequence of the target gene.
  • the second strand is from Ito 9 nucleotides shorter than the first strand.
  • the 5 '-terminal and 3 '-terminal nucleotides of the first strand are 19 nucleotides apart.
  • the 5 '-terminal and 3 '-terminal nucleotides of the second strand are 13 nucleotides apart, in certain embodiments, the 5'- terminal and 3 '-terminal nucleotides of the first strand are 19 nucleotides apart and the 5'- terminal and 3 '-terminal nucleotides of the second strand are 13 nucleotides apart.
  • the 3 '-terminal nucleotide of the second strand is complementary to a nucleotide of the first strand that is within 3 nucleotides from the 5'- terminal nucleotide of the first strand, in certain embodiments, the 3' -terminal nucleotide of the second strand i s complementary to the 5 '-terminal nucleotide of the first strand.
  • the S'-terminal nucleotide of the second strand is complementary to a nucleotide of the first strand that is 1-4 nucleotides from the S'-terminal nucleotide of the first strand.
  • the 5 '-terminal nucleotide of the second strand is complementary to a nucleotide of the first strand that is 1-2 nucleotides from the 3'- terminal nucleotide of the first, strand.
  • a least one nucleotide of the sequence of 5' overhang is selected from the group consisting of A, U, and dT.
  • the GC content of the double stranded region is 20%-70%.
  • the first strand has a length from 19-22 nucleotides.
  • the first strand has a length of 21 nucleotides. In certain embodiments, the second strand has a length of 14-16 nucleotides.
  • the first strand has a lengtli of 21 nucleotides, and the second strand has a lengtli of 15 nucleotides. In certain embodiments, the first strand has a 3'-overhang of 2-4 nucleotides. In certain embodiments, the first strand has a 3 '-overhang of 3 nucleotides. In certain embodiments, the first strand has a 5'-overhang of 3 nucleotides,
  • a duplex RNA molecule of the present disclosure can contain at least one modified nucleotide or its analogue.
  • the at least one modified nucleotide or its analogue can be a sugar-, backbone-, and/or base- modified ribonucleotide.
  • the backbone-modified ribonucl eoti de can have a modification in a phosphodiester linkage with another ribonucleotide.
  • the phosphodiester linkage is modified to include at least one of a nitrogen or a sulphur heteroatom.
  • the modified nucleotide or its analogue can be a backbone- modified ribonucleotide containing a phosphothioate group.
  • the at least one modified nucleotide or its analogue is an unusual base or a modified base.
  • the at least one modified nucleotide or its analogue comprises inosine, or a tritylated base.
  • the modified nucleotide or its analogue is a sugar- modified ribonucleotide, wherein the 2 -OH group is replaced by a group selected from H, OR, R, halo, SH, SR. NH2, NHR, NR2, or CN, wherein each R is independently Ci-Ce alkyl, alkenyl or aikynyl, and halo is F, CI, Br, or I.
  • the first strand comprises at least one deoxynueleotide. In certain embodiments, the at least one deoxynueleotide is in one or more regions chosen from 3 '-overhang, 5 ⁇ overhang, or double-stranded region. In another embodiment, the second strand comprises at least one deoxynueleotide.
  • a modulator of KRAS signaling comprises, for example, an aiRNA In certain embodiments, a modulator of KRAS signaling comprises a RAS-re!ated aiRNA In certain embodiments, a modulator of KRAS signaling comprises a KRAS-specific aiRNA.
  • a modulator of KRAS signaling comprises, for example, one or more KRAS-specific aiRNAs that target one or more KRAS activating mutations as defined herein. In certain embodiments, a modulator of KRAS signaling comprises, for example, one or more KRAS-specific aiRNAs that target KRAS transcripts having one or more KRAS activating mutations at amino acid residues G12, G13, S 17, P34, and/or Q61.
  • a modulator of KRAS signaling comprises, for example, one or more KRAS-specific aiRNAs that target both wild type and oncogenic KRAS RNA sequences.
  • Wild type KRAS-specific aiRNAs are described, for example, in the PCT International Application WO2015139044, the content of which is hereby incorporated by reference in its entirety for any purpose.
  • Exemplasy KRAS aiRNA molecules comprising a sense strand sequence, an arstisense strand sequence or a combination of a sense strand sequence and antisense strand sequence are also shown in TABLE 4.
  • AAACUCUIJAGUUUUU 633 AAAA A CUAAGAGUUU GAG 5 1
  • the RNA duplex molecule comprises a sense strand sequence selected from the group consisting of SEQ ED NOs: 320-637.
  • the RNA duplex molecule comprises an antisense strand sequence selected from the gxoup consisting of SEQ ID NOs: 638-955,
  • the RNA duplex molecule comprises a sense strand sequence selected from the group consisting of SEQ ID NOs: 320-637 and its corresponding complimentary antisense strand sequence as depicted in TABLE 4 above.
  • a KRAS RNA duplex molecule comprises a sense strand sequence that is at least e.g., 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% or more identical to a sequence selected from the group consisting of SEQ ID NOs: 320-637
  • the RNA duplex molecule comprises an antisense strand sequence that is at least, e.g., 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% * or more identical to a sequence selected from the group consisting of SEQ ID NOs: 638-955.
  • the RNA duplex molecule comprises a sense strand sequence thai is at least, e.g., 50%, 60%, 70%. 75%, 80%, 85%, 90%, 95% or more identical to a sequence selected from the group consisting of SEQ ID NOs: 320-637 and a substantially complimentary antisense strand sequence that independently is at least, e.g., 50%, 60%, 70%, 75%, 80% 85%, 90%, 95% or more identical to the corresponding antisense strand as depleted in TABLE 4 above.
  • a modulator of KRAS signaling comprises, for example, one or more aiRNAs that target one or more effectors of the RAS/RAF/MEK/ERK/FRA signal transduction pathway.
  • a modulator of KRAS signaling comprises, for example, one or more aiRNAs that target wildtype or mutant RAS GEFs.
  • a modulator of KRAS signaling comprises, for example, one or more aiRNAs that target wildtype or mutant A-RAF, B-RAF and/or C-RAF/RAF-I.
  • a modulator of KRAS signaling comprises, for example, one or more aiRNAs that target wildtype or mutant MEK 1/2.
  • a modulator of KRAS signaling comprises, for example, one or more aiRNAs that target wildtype or mutant ERK 1/2. In certain embodiments, a. modulator of KRAS signaling comprises, for example, one or more aiRNAs that target wildtype or mutant. FRA-1.
  • the present disclosure also provides a method of preparing a duplex RNA molecule of the disclosure.
  • the method comprises synthesizing the first strand and the second strand.
  • the method comprises combining the synthesized strands under annealing conditions, in certain embodiments, the duplex RNA molecule is capable of effecting KRAS silencing.
  • the RNA strands are chemically synthesized, or biologically synthesized.
  • the first strand and the second strand are synthesized separately or simultaneously.
  • the method comprises introducing at least one modified nucleotide or its analogue into the duplex RNA molecule during the synthesizing step, after the synthesizing and before the combining step, or after the combining step.
  • the present disclosure provides an expression vector for the generation of the duplex RNA in vivo.
  • the vector comprises a nucleic acid or nucleic acids encoding the duplex RNA molecule operably linked to at least one expression-control sequence, e.g. a U6 snRNA promoter or an inducible promoter, in certain embodiments, the vector comprises a first nucleic acid encoding the first strand operably linked to a first expression- control sequence.
  • the vector comprises a second nucleic acid encoding the second strand operably linked to a second expression-control sequence, in another embodiment, the vector is a viral, eukaryotic, or bacterial expression vector.
  • the present disclosure also provides a cell comprising the expression vector.
  • the cell comprises a duplex aiRNA molecule, e. g. a KRAS-specific aiRNA
  • the present disclosure further provides a method of modulating KRAS signaling.
  • the method comprises administering an. asymmetrical duplex RNA. molecule of the disclosure, in certain embodiments, the method comprises administering an asymmetrical duplex RNA molecule of the disclosure in an amount effective to silence KRAS expression or otherwise reduce KRAS expression. ID certain embodiments, the method is in a cell or an organism.
  • the method comprises contacting said cell or organism with an asymmetrical duplex RNA molecule of the disclosure, for example, under conditions wherein selective KRAS gene silencing can occur, in certain embodiments, the method comprises mediating a selective KRAS gene silencing affected by the duplex RNA molecul e towards KR AS or nucleic acid having a sequence portion substantially corresponding to the double-stranded RNA.
  • said contacting step comprises introducing said duplex RNA molecule into a target cell in culture or in an organism in which the selective KRAS silencing can occur.
  • the introducing step is chosen from transfection, lipofection, electroporation, infection, injection, oral administration, inhalation, topical administration, or regional administration.
  • the introducing step comprises using a pharmaceutically acceptable excipient, carrier, or diluent.
  • the pharmaceutically acceptable excipient, carrier, or diluent is chosen from a pharmaceutical carrier, a positive- charge carrier, a liposome, a protein carrier, a polymer, a nanoparticle, a nanoemulsion, a lipid, and a lipoid.
  • a modulator of KRAS signaling may comprise one or more inhibitors that target the KRAS activation of the RAF/MEK ERK ' FR A- 1 signaling pathway.
  • the method comprises administering an asymmetrical duplex RNA molecule of the disclosure in an amount effective in reducing (e.g., silencing) the KRAS activation of the RAF/MEK/ERK FRA- 1 signaling pathway.
  • a modulator of KRAS signaling may comprise one or more inhibitors that target the oncogenic KRAS activation of the RAF MEK/ERK/FRA ⁇ 1 signaling pathway.
  • the method comprises administering an asymmetrical duplex RNA molecule of the disclosure in an amount effective in reducing (e.g., silencing) the oncogenic KRAS activation of the RAF/MEK/ERK/F A- 1 signaling pathway.
  • a modulator of KRAS signaling may comprise an inhibitor of RAF activity (e.g. A-RAF, B-RAF, RAF1). in certain embodiments, a modulator of KRAS signaling may comprise an inhibitor of mitogen-activated kinase activity (e.g., MEKl, MEK2), an inhibitor of ER activity (e.g., ERKl, ERK2). In certain embodiments, a modulator of KRAS signaling may comprise inhibitor of FRA-1 activity or any combination thereof.
  • RAF activity e.g. A-RAF, B-RAF, RAF1
  • a modulator of KRAS signaling may comprise an inhibitor of mitogen-activated kinase activity (e.g., MEKl, MEK2), an inhibitor of ER activity (e.g., ERKl, ERK2).
  • a modulator of KRAS signaling may comprise inhibitor of FRA-1 activity or any combination thereof.
  • downstream targets may also be suitable targets for an inhibitor of KRAS signal
  • a modulator of KRAS signaling may comprise, for example, an RNA. interfering agent that inhibits one or more downstream effectors of KRAS signaling.
  • a modulator of KRAS signaling m )' comprise an inhibitor of A-RAF, B-RAF, and/or C-RAF, for example, an A-RAF, B-RAF, and/or G-RAF RNA interfering agent (e.g., see WO2009143372, the content of which is hereby incorporated by reference in its entirety for any purpose).
  • a modulator of KRAS signaling may comprise an inhibitor of MEK activity, for example, a MEK RNA interfering agent (e.g., see published Patent Application No. 2009/0239936, the content of which is hereby incorporated by reference in its entirety for any purpose).
  • a MEK RNA interfering agent e.g., see published Patent Application No. 2009/0239936, the content of which is hereby incorporated by reference in its entirety for any purpose.
  • a modulator of KRAS signaling may comprise an inhibitor of ERK activity, for example, a ERK RNA interfering agent (e.g., see published Patent Application No. 2009/0239936, the content of which is hereby incorporated by reference in its entirety for any purpose).
  • a ERK RNA interfering agent e.g., see published Patent Application No. 2009/0239936, the content of which is hereby incorporated by reference in its entirety for any purpose.
  • a modulator of KRAS signaling may comprise an inhibitor of
  • FRA-1 activity for example, a FRA-1 RNA interfering agent (e.g. see U.S. Patent No. 6,124,133, the content of which is hereby incorporated by reference in its entirety for any purpose).
  • a FRA-1 RNA interfering agent e.g. see U.S. Patent No. 6,124,133, the content of which is hereby incorporated by reference in its entirety for any purpose.
  • a modulator of KRAS signaling may comprise an antigen- binding protein or fragment thereof (e.g. a recombinant antigen-binding protein) that targets RAS protein, e.g. an oncogenic KRAS (e.g. see U.S. Patent Nos. 4,820,631; 5,084,380, 4,898,932; 5,081,230; 5, 1 12,737; 5,028,527, the contents of which, are hereby incorporated by reference in their entireties for any purpose), RAF protein (e. g. See WO2014047973, the content of which is hereby incorporated by reference in its entirety for any purpose), MEK protein, ERIC protein and/or FRA- 1 protein.
  • RAS protein e.g. an oncogenic KRAS (e.g. see U.S. Patent Nos. 4,820,631; 5,084,380, 4,898,932; 5,081,230; 5, 1 12,737; 5,028,527, the contents of which, are hereby incorporated by reference
  • a modulator of KRAS signaling may comprise a small molecule inhibitor of KRAS signaling (see, for example, WO2016123378A1, the content of which is hereby incorporated by reference in its entirety for any purpose and TABLE 5 below).
  • a modulator of KRAS signaling may comprise a covalent inhibitor of KRAS G12C (see for example, WO2014152588 and WO2016049524, the contents of which are hereby incorporated by reference herein in their entireties for any purpose).
  • a modulator of KRAS signaling may comprise an inhibitor of prenyl transferase activity
  • a modulator of KRAS signaling comprises one or more agents each independently chosen from inhibitors of farnysyltransferase or geranylgeranyl transferase I (e.g. , see U.S. Patent No. 5,965,539, the content of which is hereby- incorporated by reference in its entirety for any purpose).
  • a modulator of KRAS signaling may comprise, for example, a small molecule RAF inhibitor.
  • a small molecule RAF inhibitor can inhibit the RAF--MEK---ERK pathway signal ing in cells expressing the BRAFV600E oncogene.
  • Exemplary BRAF inhibitors include, but are not limited to, vemurafenib (RG7204 or PLX4032) which has been approved by the FDA for treatment of melanoma., dabrafenib which has been approved by FDA for treatment of cancers associated with a mutated version of the gene BRAF, GDC-Q879, PLX-4720 (Plexxikon Roche) (R72Q4), Sorafenib Tosylate, dabrafenib and/or LGX818 or any combination thereof.
  • a modulator of KRAS signaling may comprise, for example, a small molecule MEK inhibitor.
  • a MEK inhibitor may comprise, for example, an ATP-competitive MEK inhibitor, a non- ATP competitive MEK inhibitor, and/or an ATP- unc o mpet i t i ve MEK inliibiior.
  • Exemplary MEK inhibitors include, but are not limited to, trametinib (GSK1120212; JTP 74057), for treatment of BRAF-mutated melanoma and possible combination with BRAF inhibitor dabrafenib (GSK2118436) to treat BRAF-mutated melanoma; selumetmib (ARRY- .
  • NSCLC non-small cell lung cancer
  • binimeti ib MEK162, ARRY- 162, ARR Y -43 162
  • PD-3259Q1 for breast cancer, colon cancer, and melanoma
  • Cobimetinib GDC-0973; XLS 18
  • AS703026 primasertib
  • the modulator of KRAS signaling may comprise, for example, RG7304, a small molecule MEK inhibitor with a structure based on a coumarin skeleton. It selectively inhibited RAFl (C-RAF), B-RAF, mutant B-RAF (V600E), and MEKl In in vitro studies and showed a strung and broad spectrum of antitumor activities both in vitro in various tumor cell lines and in vivo in mouse xenograft models.
  • C-RAF C-RAF
  • B-RAF B-RAF
  • V600E mutant B-RAF
  • a modulator of KRAS signaling may comprise, for example, a non-ATP-competitivc small- molecule MEK inhibitor (e.g. PD 098059, U0126, PD 184352 and its derivatives) or a biological inhibitor (e.g. anthrax lethal toxin and Yersinia outer protein J).
  • a modulator of KRAS signaling may comprise, for example, a pyrrole derivative of MEK kinase.
  • a modulator of KRAS signaling may comprise, for example, a 4-anilino-3-cyano-6,7-dialkoxyqumoline, including 4-anilino-3- cyano-6,7-dialkoxyquinolines (e.g., see WO2013059320, the content of which is hereby incorporated by reference in its entirety for any purpose).
  • a modulator of KRA S signaling may comprise, for example, a small molecule ERIC inhibitor.
  • exemplary ERK inhibitors include, but are not limited to, SCH772984 which is an ATP-cotnpetitive ER l and ERK2 inhibitor, MK-8353/SCH900353, a clinical grade analogue of SCH772984, which is currently being tested in Phase I clinical trials, BVD-523 (Biomed Valley Discoveries) and RG7842 (GDCQ994; Genentech/Roche) which is a selective inhibitor of ERK 1/2. Phase I clinical trials evaluating RG7842 as a single agent or in combination with cobirnetinib in solid tumors are ongoing.
  • SCH772984 studies conducted with SCH772984 indicate that the small molecule can inhibit cellular proliferation and cause apoptosis selectively in tumor cell lines that carry RAS or BRAF mutations, and also induce significant tumor regressions in mice with BRAF- or RAS-mutant xenografts. SCH772984 also demonstrated inhibition activity in cells that were resistant to either BRAF or MEK inhibitors and in cells that became resistant to the dual combination of these inhibitors.
  • a modulator of KRAS signaling may comprise, for example, a small molecule inhibitor with a thienyl benzenes ulfonate scaffold that can inhibit ERK 1/2 substrates containing an F-site or DEF (docking site for ERK, FXF) motif (e.g. see U.S. Patent No. 9, 115,122, the content of which is incorporated by reference herein in its entirety for any purpose).
  • a modulator of KRAS signaling may comprise, for example, an inhibitor of ERK2-mediated phosphorylation of c-FOS, e.g. SF-3-026 and its analogues.
  • the modulator of KRAS signaling may comprise, for example, an inhibitor of AP-.1 promoter activity in cells with constitutively active ERK 1/2, e.g. SF-3-030 and its analogues.
  • exemplary small molecule inhibitors of ERKl/2-regulated proteins include, but are not limited to the compounds depicted in TABLE 6.
  • a modulator of RAS signaling may comprise, for example, an antigen-bi ding protein or fragment thereof (e.g. a recombinant antigen-binding protein) that targets RAS protein,
  • the RAS protein is chosen from an oncogenic KR AS (e.g., see U.S. Patent Nos.
  • RAF protein e.g., see WO2014047973, the content, of which is hereby incorporated by reference in its entirety for any purpose
  • MEK protein e.g., WO2014047973, the content, of which is hereby incorporated by reference in its entirety for any purpose
  • MEK protein e.g., WO2014047973
  • ERK. protein e.g., ERK. protein and/or FRA-1 protein.
  • a modulator of KRAS signaling may comprise, for example, a peptide that binds to and inhibits the activity of RAS protein.
  • the RAS protein is chosen from RAF protein, MEK protein, ERK protein and/or FRA-1 protein, in certain embodiments, the peptide can be, for example, a chimeric peptide comprising a cell penetrating peptide, e.g. pro-apoptotic RAS and/or RAF peptides (see e.g. WO2015001045, the content of which is hereby incorporated by reference in its entirely for any purpose).
  • compositions comprising a modulator of KRAS signaling.
  • the composition is for the treatment of a KRAS-assoeiated disorder or disease, e.g. cancer.
  • the composition comprises a therapeutic agent .
  • the therapeutic agent can be, for example, a chemotherapeutic agent, a targeted agent, or an immunotherapeutic agent.
  • a modulator of signaling may be combined, lor example, with an immunotherapeutic agent, e.g., any agent that can induce, enhance, or suppress an immune response in a subject,
  • an immunotherapeutic agent comprises an antibody or a recombinant antigen-binding protein or fragment thereof, in certain embodiments, a recombinant antigen-binding protein, or fragment thereof, can be, for example, monospecific, bispecific or multi-specific and monovalent or bivalent recombinant antigen-binding protein.
  • an antigen-binding protein can be an asymmetric bispecific antibody, an asymmetric bispecific IgG4, a CrossMab binding protein, a DAI 7 (dual action Fab antibody; two-in-one), a DM (dual action Fab antibody; four-in-one), a DutaMab, a DT-IgG, a knobs ⁇ 1 ⁇ 2 ⁇ holes binding protein, a Charge pair binding protein, a Fab-arm exchange binding protein, a SEEDbody, a Triomab (Triomab quadroma bispecific or removab bispecific), a LUZ-Y, a Fcab, a ⁇ -body, an iMab (innovative multimer), an Orthogonal Fab, a DVD-Ig binding protein, an IgG(H)-scFv, an scFv-(H)IgG, an IgG(L)-scF
  • a bispecific tandem nanobody a bispecific trivalent tandem nanobody, a nanobody- HSA, a BiTE (bispecific T-cell engager) binding protein, a Diabody, a DART (dual affinity retargeting) binding protein, a TandAb (tetravalent bispecific tandem antibody), an scDiabody, an scDiabody-CH3, a Diabody-CH3, a Triple Body, a Miniantibody, a Minibody, a TriBi.
  • an scFv-CFB ⁇ an scFv-CFB ⁇ , a Fab-scFv, an scFv-CH-CL-scFv, a F(ab')2, a F(ab')2 scFv2, an scFv-KIH, a Fab-scFv-Fc, a Tetravalent HCAb, an scDiabody-Fc, a Diabody-Fc, a Tandem scFv-Fc, a Fabsc, a bsFc-1/2, a CODV-Ig (cross-over dual variable immunoglobulin), a hiclonies antibody, an Inlrabody, a Dock and Lock binding protein, an ImmTAC, an HSAbody, an scDiabody- HS A, a Tandem scFv- ⁇ , an IgG-IgG binding protein, a Cov-X-Body, and/or an s
  • an immunotherapeutic agent specifically binds to a specific cytokine, cytokine receptor, co-stimulatory molecule, co-inhibitory molecule, or immunomodulatory receptor that modulates the immune system.
  • an immunotherapeutic agent .specifically binds to a component of a regulatory T cell, myeloid suppressor cell, or dendritic cell.
  • an immunolherapeuti c agent can be a cytokine, for example, an interferon (IFN), interleukin, or the like.
  • an immuno therapeutic agent can be interferon (IFNa or ⁇ ), type 2 (IFNy), or type III (IFNX).
  • An immunothsrapeuiic agent can also be interleukin- 1 (JL-1), interieukin- 1 a (IL- la), interleukin- 1 ⁇ (IL- ⁇ ), interleukin-2 (IL- 2), interleukin-3 (11,-3), interleukin-4 (1L-4), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin- 10 (IL-10), interleukin- .1 1 (EL- 11), interieukin- 12 (IL-12), interleukin- 13 (IL-13), or interleukin- 18 (EL- 18), or the like.
  • a modulator of KRAS signaling described herein may be combined, for example, with an immunotherapeutic agent.
  • the immunotherapeutic agent targets and/or binds a cancer or tumor cell marker or component.
  • Exemplary cancer or tumor cell markers or components include , but are not limited to, are not limited to, epidermal growth factor receptor (EGFR; EGFR ; ErbB-1; HERE); ErbB-2 (HER2/neu); ErbB-3/HER3; EAB-4/HER4; EGFR ligand family; insulin-like growth factor receptor (IGFR) family; IGF-binding proteins (IGFBPs) IGFR ligand family (IGF-IR); platelet derived growth factor receptor (PDGFR) family; PDGFR ligand family; fibroblast growth factor receptor (FGFR) family: FGFR ligand family; vascular endothelial growth factor receptor (VEGFR) family; VEGF family; HGF receptor family; TRK receptor family; ephrin (E
  • TNF tumor necrosis factor
  • TNF tumor necrosis factor
  • T ' NTRSF tumor necrosis factor receptor superfamily
  • TRAIL- receptor cancer-testis (CT) antigens; lineage-specific antigens; differentiation antigens; alpha-actinin-4 ARTCl ; breakpoint cluster region- Abelson (Bcr-Ahl) fusion products: B- RAF; caspase-5 (CASP-5); caspase-8 (CASP-8); beta-catenin (CTNNBl); cell division cycle 27 (CDC27); cyclin- dependent kinase 4 (CDK4); CDK 2A; CO A- 1; dek-can fusion protein: EFTUD-2; Elongation factor 2 (ELF2); Ets variant gene 6/aeute myeloid leukemia 1 gene ETS (ETC6-AML1) fusion protein; fibronectin (FN); GPNMB; low density lipid receptor/GDP-L
  • M-H -I; LAGE; LAGE-1; CTL-recognizsd antigen on melanoma CAMEL
  • MAGE-A1 MAGE-1) MAGE-A2; MAGE- A3; MAGE-A4; MAGE- AS; MAGE-A6; MAGE-A8; MAGE-A9; MAGE- A 10; MAGE- Al J ; MAGE-A12; MAGE- 3; MAGE-B !; MAGE-B2; MAGE-B5; MAGE-B6; MAGE-C1; MAGE-C2 mucin 1 (MUC1); MART-l Melan-A (MLA A); gplOO; gplOO/Pmel 17; tyrosinase (TYR); TRP-1; HAGE; NA-88; NY-ESO-1; N Y-ES O-l L AGE-2: SAGE; Sp!7; SSX- 1; 2; 3; 4; TRP2-1NT2; carcinoe
  • a modulator of RAS signaling described herein may be combined, for example, with one or more immunotherapeutie antibodies, each independently chosen from trastuzumab (antj- HER2/neu antibody); pertuzumab (anti-HER2 inAb); celuximab (chimeric monoclonal antibody to epidermal growth factor receptor EGFR); panitumumab (anti-EGFR antibody); nimotuzumab (anti-EGFR antibody); za!utumumab (anti- EGFR mAb); necitumumab (anti- EGFR mAb); MDX-210 (humanized anti-HER-2 bispecifie antibody); MDX-210 (humanized anti-HER-2 bispecifie antibody); MDX-447 (humanized anti-EGF receptor bispecifie antibody); rituximab (chimeric murine/human- anti-CD20 mAb); obinutuzumab (anti-CD20 mAb); ofalumumab (anti-HER2/neu antibody
  • BCG oncolym
  • SMART M195 Ah humanized 13' 1 LYM-1 (Oncolym)
  • Ovarex B43. 13, anti-idiotypic mouse mAb
  • Zenapax SMART Anti-Tac (IL- 2 receptor); SMART Ml 95 Ab, humanized Ab, humanized
  • ovoMAb ⁇ G2 pancarcinoma specific Ab
  • TNT chimeric mAb to histone antigens
  • TNT chimeric mAb to hi stone antigens
  • Gliomab-H Monoclonal s— Humanized Abs
  • EMD-72000 chimeric-EGF antagonist
  • LymphoCide humanized IL.L.2 antibody
  • MDX-260 bispecifie, targets GD-2, ANA Ab, SMART ID
  • an i mmun otherapeutic agent can be a cell, for example, an immune cell.
  • an immune cell particularly one that is specific to a tumor, can be activated, cultured, and administered to a patient.
  • the immune cell can be a natural killer cell, lymphokine-aciivated killer cell, cytotoxic T-cell, dendritic cell, or a tumor infiltrating lymphocyte (TIL).
  • TIL tumor infiltrating lymphocytes
  • tumor infiltrating lymphocytes refers to white blood cells ((i.e., T cells, B cells, N cells, macrophages) that have left the bloodstream and migrated into a tumor.
  • an imr inotherapeutic agent can be, for example, sipuleucel-T (Provenge).
  • a modulator of KRAS signaling as disclosed herein may be combined with an inhibitor of an immune checkpoint.
  • Immune checkpoint proteins can regulate and maintain self-tolerance and the duration and amplitude of physiological immune responses including cytotoxic T cell activity.
  • Non-limiting examples of immune checkpoint proteins include cytotoxic T- lymphocyte-associated antigen (CTLA, for example, CTLA4) and its ligands CD 80 and CDS 6; programmed cell death protein (PD, for example, PD-1) and its ligands PD-Ll and PDL2; indoleamine-pyrrole 2,3-dioxygenase-l (IDOl); T cell membrane protein (TIM, for example, TIM3); adenosine A2a receptor (A2aR); lymphocyte activation gene (LAG, for example, LAGS); killer immunoglobulin receptor (KIR); or the like.
  • CTLA cytotoxic T- lymphocyte-associated antigen
  • PD programmed cell death protein
  • IDOl indoleamine-pyrrole 2,3-dioxygenase-l
  • T cell membrane protein TIM, for example, TIM3
  • A2aR lymphocyte activation gene
  • LAG killer immunoglobulin receptor
  • KIR killer immunoglobul
  • immune checkpoint inhibitor refers to a molecule that can completely or partially reduce, inhibit, interfere with, or modulate one or more immune checkpoint proteins that regulate T ⁇ cell activation or function.
  • immune checkpoint inhibitor can refer to a molecule that can interfere or/and prevent the interaction of PD- 1 with its !igand, either PD-Ll or PD-L2.
  • the immune checkpoint inhibitor can target CTLA4. In certain embodiments, the immune checkpoint inhibitor can target PD-1. In certain embodiments, the immune checkpoint inhibitor can target PD-Ll. In certain embodiments, the immune checkpoint inhibitor can target PD-L2. In certain exnbodiments, the immune checkpoint inhibitor can target LAG 3. in certain embodiments, the immune checkpoint inhibitor can target B7-H3. In certain embodiments, the immune checkpoint inhibitor can target B7-H4. In certain embodiments, the i mmune checkpoint inhibitor can target TIMS.
  • the immune checkpoint inhibitor can be a small molecule.
  • the immune checkpoint inhibitor can be a small molecule that, competes with an antibody or other antigen-binding protein, or fragment thereof, for binding to an immune checkpoint molecule.
  • the immune checkpoint inhibitor can be a small molecule that competes with an anti-PD- 1 antibody, e.g., nivolumab, pembrolizumab, pidilizumab, BMS 936559, or atezolizumab as disclosed herein, for binding to PD- 1.
  • the immune checkpoint inhibitor can be a small molecule that competes with an anti-PD-Ll antibody, e.g., atezolizumab, avelumab, or durvahimab as disclosed herein, for binding to PD-L1.
  • the immune checkpoint inhibitor can be a small molecule that competes with an anti-PD-L2 antibody, e.g., rHIgM12B7 or Dana-Farber patent anti-PD-L2 as disclosed herein, for binding to PD-L2.
  • an anti-PD-L2 antibody e.g., rHIgM12B7 or Dana-Farber patent anti-PD-L2 as disclosed herein, for binding to PD-L2.
  • the immune checkpoint inhibitor can be a small molecule that competes with an anti-CTL4 antibody, e.g., ipilimumab, tremelimumab or AGEN1884 as disclosed herein, for binding to CTL4.
  • an anti-CTL4 antibody e.g., ipilimumab, tremelimumab or AGEN1884 as disclosed herein, for binding to CTL4.
  • the immune checkpoint inhibitor can be a small molecule that competes with an anti- VISTA antibody, e.g., the Janssen patent anti-VISTA antibody, Igenica patent anti-C10orf54 antibody . or the Amplimmune patent anti-B7-H5 antibody as disclosed herein, for binding to the immune checkpoint protein, VIST
  • the immune checkpoint inhibitor can be a small molecxile that rescues the inhibition of cell proliferation by recombinant PD-L1 in a mouse splenocyte assay by about 10% to about 95% (the mouse splenocyte assay is described in detail in, for example, WO2016142833).
  • the immune checkpoint inhibitor can be, for example, a 1,2,4- oxadiazole and thiadiazole compound, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  • the immune checkpoint inhibitor can suppress or inhibit the programmed cell death 1 (PD-1) signaling pathway in T cells.
  • the immune checkpoint inhibitor can be, for example, a compound of formula (I)
  • X is O or S
  • Ri and R2 independently are a side chain of an amino acid or hydrogen.
  • heterocyclyl and heteroaryl are further substituted by one or more substituents such as hydroxy, alkoxy, halo, arnino, nitro, cyano or alky! and optionally wherein two or three carbon atoms of the (Ci-Cfi)alkyl ? (C2-C6)alkenyl or (C2-G>)alkynyl form part, of a 3-7-membsred carbocyclic or heterocyclic ring (such as a cyclobutyl. or oxirane ring);
  • R3 is hydrogen, -CG-[Aaal] m , [Aaal.] m , [Aaal] m -CO-[Aa.a l] m , -S(0) P -[Aaal] m , - CONR-Rs, -CORe, -SQiRc, (Ci-Cejalkyl, (C2-Gs)alkenyl or (Cs-C ⁇ alkynyl; wherein (Ci- C6)alkyL (C2-C6)alkenyl and (C2-C6)alkynyl are optionally substituted by one or more substituents selected from amino, alkylamino, acylamino, -COO-alkyl, carboxylic acid, carboxylate, thiocarboxylate, thioaeid, -CONRTRS, hydroxy, aryl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl, (cyclo
  • R4 and R5 independently are hydrogen or absent
  • Re is hydrogen, alkyl, alkenyl, alkynyi, aralkyl, aryl, heteroaralkyl, heteroaiyl, cycloalkyl, (cycloalkyl )alkyl, amino, aminoalkyl, hydroxyalkyl, alkoxyalkyl, acyl, [Aaa2]n, - CO[Aaa2]n, [Aaa2]n-CO-[Aaa2] n or -S(0) P -[Aaa2]n;
  • R.7 and Rs independently are hydrogen, (Ci-C ⁇ >)alkyl, (C2-Ce)alkenyl, (C2-C3 ⁇ 4)alkynyL aryl or heterocyclyl; wherein (Ci-Cs)alkyi, (C2-C6)alkenyl, (C2-C6 ' )alkynyl, aryl and heterocyclyl are optionally substituted by one or more substituents selected from halogen, hydroxy!, amino, nitro, cyano, cycloalkyl, heterocyclyl.
  • heteroaryl aryl, guanidino, (ey oalkyl)alkyl, (heterocyclyl)alkyl and (heteroaryl)alkyl; optionally wherein two or thrse carbon atoms of the (Ci-C6)a1kyl, (C2-C6)alkenyl or (C2-Ce)alkynyl form part of a 3-7- membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring);
  • R? and Rs together with the nitrogen to which they are attached form an optionally substituted 3-7-membered ring containing 0-2 additional heteroatoms independently selected from N, O and S in any stable combination; wherein the optional substituent at each occurrence is selected from hydroxyl, -COOH, -CQO-aikyl, amide, halo, amino, nitro and eyano;
  • R 3 is hydrogen or alkyl. alkenyl. aikynyl, acyl, aralkyL aryl, heteroaralkyl, heteroaryl, cycloalkvl, (cycloalkyi )alkyl. aminoalkyl, hydroxy alkyl or alkoxyalkyl;
  • Rb is hydrogen., alkyl, alkenyl, aikynyl, acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkvl, (cycloalkyl)alkyl, aminoalkyl, hydroxyalkyl or alkoxyalkyl; or Rb and R 2 , together with the atoms to which they are attached, may form pyrrolidine or piperidine optionally substituted with one or more groups independently selected from hydroxy, halo, amino, eyano and alkyl;
  • R is (Ci-Ce)alkyl, cycloalkvl, aryl, heterocyclcyl or heteroaryl; wherein the said (Ci- C(5)alkyi, cycloalkyi aryl, heterocyclcyl or heteroaryl is optionally substituted by one or more substituents selected from carboxylic acid, hydroxy, alkyl, alkoxy, amino, alkylamino, acylamino, carboxylic ester, cycloalkyi, heterocyclyL heteroaryl, (cycloalkyl)alkyL (helerocyclyl)alkyl or (heieroaryl)aikyl;
  • n and n independently are integers selected from 1 to 3;
  • p is an integer selected from 1 to 2;
  • Ri is not a side chain of Ser or Thr
  • R2 is a side chain of Asp
  • Asa Glu or Gin
  • R3 is hydrogen
  • Re is hydrogen
  • Ra and Rfi are hydrogen.
  • compositions and methods of using and making the compounds of Formula (I) are disclosed in WO2016142833 (e.g. Compound Nos. 1- 124 of WO2016 42833), the content of which is hereby incorporated by reference herein in its entirety.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of t! modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of a compound of Formula (I).
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • the method is effective at. sensitizing tumor cells to a compound of Formula (I).
  • the method changes (e.g., enhances) the efficacy of a compound of Formula (I).
  • the tumor cells are resistant to treatment with a compound of Formula (I) alone.
  • the compound of Formula (I) can be, for example.
  • the compound of Formula (I) can be, for example, Compound No. 14 o
  • the compound of Formula (I) can be, for example, Compound No, 60 of WO2016142833 havin the structure of:
  • the compound of Formula (I) can be, for example, Compound No, 75 of WO2016142833, havin the structure of:
  • the immune checkpoint inhibitor can be, for example, a 3 ⁇ substituted 1,3,4-oxadiazo!e and thiadiazole compound, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  • the immune checkpoint inhibitor can be, for example, a compound of formula (II):
  • X is O or S
  • each dotted line [ ] independently represents an optional bond
  • Ri is hydrogen or -CO-Aaa
  • Aaa represents an amino acid residue
  • R.2 is side chain of an amino acid, hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, heteroaraikyi, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxy!
  • Ra independently for each occurrence, is alkyl, alkoxy, halo, hydroxy!, amino, - C(0)OH, aralkyl, aryl, a!koxy, heteroaralkyl, heteroaryl, cycioalkyl., (cyeloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyk or any two Ra groups attached to the same carbon atom together represent an oxo ( ⁇ 0) or tJhioxo ( :;; S);
  • each of R4 and Rs independently is hydrogen or absent
  • R0 is hydrogen or alkyl.
  • compositions and methods of using and making the compounds of Formula (II are disclosed in WO2016142894 (see, for example. Compound Nos, 1- 30 of WO2016142894) and WO2016142886 (see, for example, Compound Nos. 1-62 of WO2016142886), the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor compri sing administering an effective amount of a modulator of RAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of a compound of Formula (II).
  • the method is effective at sensitizing tumor cells to a compound of Formula (II).
  • the method changes (e.g., enlianc.es) the efficacy of a compound of Formula ( ⁇ ).
  • the tumor cells are resistant to treatment with a compound of Formula (II) alone.
  • the compound of Formula (II) can be, for example, Compound No. 11 of WO2016142894, having the structure of:
  • the compound of Fornmla (II) can be, for example.
  • Compound No. 13 of WO2016142894 having the structure of:
  • the compound of Formula (II) can be, for example, Compound No. 32 of O2016142886, having the structure of:
  • the compound of Formula (II) can be, for example, Compound No. 43 of WO2016142886, havin the structure of:
  • the immune checkpoin inhibitor can be, for example, a 1,3,4- oxadiazols and thiadiazole compound or a stereoisomer thereof or a pharmaceutically acceptable salt thereof that can suppress or inhibit the programmed cell death 1 (PD- 1) signaling pathway.
  • PD- 1 programmed cell death 1
  • the immune checkpoint inhibitor can be, for example, a compound of formula (III):
  • each dotted line [- - - - ] independently represents an optional bond;
  • X is O or S;
  • Ri and R2 independently are a side chain of an amino acid or hydrogen.
  • cycloalkyl, aryl, heterocyclyl and heteroaryl are further substituted by one or more substituents such as hydroxy, alkoxy, halo, amino, nitro, cyano or alkyl and optionally wherein two or three carbon atoms of the (Ci -GOalk l, (Ci- C6)alkenyl or (C2-C6)alkynyl form part of a 3-7-membered carbocyciic or heterocyclic ring (such as a cyelobutyl or oxirane ring);
  • R3 is hydrogen, -CO-[Aaa l]m, [Aaaljm, [Aaall]m-CO-[Aaal]m, -8(0 )p-[ Aaaljm, - CONR7R8, -CORc, -SChRc, (Ci-C «)alkyL (C 2 -C ⁇ s)alkenyl or (C2-Gs)alkynyl; wherein (Ci- Ce)alkyl, (C2-C6)alkenyl and (C2 ⁇ Ce)aJkynyl are optionally substituted by one or more substituents selected from amino, alkylamino, acylamino, -COO-alkyl, carboxylic acid, carboxylate, thiocarboxylate, thioacid.
  • - CO R7R8 hydroxy, aryl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl, (cycloalkyl)alkyl.
  • R.4 and R5 independently are hydrogen or absent
  • R ⁇ 5 is hydrogen, alkyl, alkenyl, alkynyl, aralkyl, ary!, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl )alkyi, amino, aminoalkyl, hydroxyalkyl, alkoxyalkyl, acyl, [Aaa2]n, - CO- 1 Aaa2Jn, [Aaa2]n-CO-[Aaa2jn or -S(0)p-[Aaa2] n ;
  • R? and Rs independently are hydrogen, (Ci ⁇ Cs)alky1, (C2-C6)alkenyl, (C2-C6)alkynyl, aryl, cycloalkyl or heterocyclyl; wherein (Ci-Ce)alkyL (C2-C «)aikenyl and (C2-Ce)alkynyl, aryl and heterocyclyl axe optionally substituted by one or more substituents selected from halogen, hydroxy!, amino, nitro, cyano, cycloalkyl, heterocyclyl, heteroaryl, aryl, guanidino, (cycloalkyl)alkyi, (heterocyclyl)alkyl and (heteroaryl)aikyl; optionally wherein two or three carbon atoms of the (Ci-Cs)alkyl, (C2-Ce)alkenyl or (C2-Ce)alkynyl form part of a 3-7- membered carbocyclic or
  • R? and Rs together with the nitrogen to which they are attached form an optionally substituted 3-7-membered ring containing 0-2 additional heteroatoms independently selected from N, O and S in any stable combination; wherein the optional suhstituent at each occurrence is selected from hydroxyl, -COOH, -COOalkyl, amide, halo, amino, nitro and cya.no;
  • Ra is hydrogen or alky , alkenyl, alkynyl., acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, aminoalkyl, hydroxyalkyi or alkoxyalkyl; or Ra and R2, together with the atoms to which they are attached, form hetcrocycloatkyl ring optionally substituted with one or more groups independently selected from hydroxy!, halo, amino, cyano and alkyl;
  • Rb is hydrogen or alkyL alkenyl, alkynyl, acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyL aminoalkyl, hydroxyalkyi or alkoxyalkyl;
  • Re is (Ci-C6)alkyi, cycloalkyl, aryl, heterocyclyl or heteroaryl; wherein the said (Ci- Cs)aikyL cycloalkyl, aryl, heterocyclyl or heteroaryl is optionally substituted by one or more substituents selected from carboxyiic acid, hydroxy, alkyl, alkoxy, amino, alkylamino, acylamino, carboxyiic ester, cycloalkyl, heterocyclyl, heteroaryl, (cycloalkyl)alkyL (heterocyclyl)alkyl or (heteroaryl)alkyl;
  • n and n independently are integers from 1 to 3;
  • p is an integer selected from 1 to 2;
  • Ri is not. a side chain of Ser, Thr, Phe, Ala or Asn, when R-i is side chain of Ser, Ala, Glu, Gin, Asn or Asp, R3 is hydrogen, -CO-Ser, -CO-Thr or -CO- Asn and Ra, Rb and Re are hydrogen.
  • compositions and methods of using and making the compounds of Formula (1) are disclosed in WO2016142852 (e.g. Compound Nos. 1-55 of WO2016142852), the content of which is hereby incorporated by reference herein in its entirety.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against, a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of a compound of Formula (III).
  • the method is effective at sensitizing tumor cells to a compound of Formula (III).
  • the method changes (e.g., enhances) the efficacy of a compound of Formula (III).
  • the tumor cells are resistant to treatment with a compound of Formula (III) alone.
  • the compound of Formula (III) can be, for example, Compond No. 1 havin the structure of:
  • the immune checkpoint inhibitor can be, for example, a cyclic compound of formula (TV) or a stereoisomer thereof or a pharmaceutically acceptable salt thereof that can suppress or inhibit the programmed cell death 1 (PD- 1) signaling pathway.
  • TV cyclic compound of formula
  • PD- 1 programmed cell death 1
  • the immune checkpoint inhibitor can be, for example, a compound of formula IV):
  • X is Cm, O, NH or S
  • Rj , R2 and R$ independently are a side chain of an amino acid, hydrogen, (C_-C «)aikyl,
  • heterocyclyl aikyL (heteroaryl)alkyl, -SH and -S- (alkyl); optionally wherein cycloalkyl, aryl, heterocyclyl and heteroaiyl are further substituted optionally by one or more substituents such as hydroxy, alkoxy, halo, amino, nitro, cyano or alkyl; optionally wherein two or three carbon atoms of the (Ci -C6)alkyL (C2-C6)aikenyl or (C2- Cs)alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyciobutyl or oxirane ring);
  • Ri', Ri Ri and R5 independently are hydrogen or alkyl
  • Ri and Ri' together with the carbon atom to which they are attached, may optionally form an optionally substituted cycloalkyl or heterocycloalkyl ring;
  • Ri and R together with the atoms to which they are attached, may optionally form a heterocyclic ring optionally substitated with one or more groups independently selected from amino, cyano, alkyl, halo and hydroxy;
  • R2 and R?.' together with the carbon atom to which they are attached, may optionally form a optionally substituted cycloalkyl or heterocycloalkyl ring;
  • R2 and R. together with the atoms to which they are attached, may optionally form a heterocyclic ring optionally substituted with one or more groups independently selected from amino, cyano, alkyl, halo and hydroxy;
  • RA and R4' independently are hydrogen or alkyl
  • Ra and Ra' are each hydro •;gen; or together represent an oxo (-O) group
  • Rb and Rb ' are each hydrogen; or together represent an oxo ( ⁇ C)) group; Rc at each occurrence is independently hydrogen or lkyl;
  • Rd is amino or - H-C(0)"(CH 2 )r ⁇ CH3
  • n is an integer from 0 to 3;
  • thai Rs is not a side chain of Ser, Asp, Ala, He, Phe, Trp, Lys, Glu and Thr, when Ri is a side chain of Ala, Ser, Thr or Leu, I1 ⁇ 2 is a side chain of Asp, Asn, Glu or Gin and R; and c are hydrogen.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of RAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount, of a modulator of KRAS signaling, e. g., a KRAS aiRNA, as defined herein, combined with an effective amount of a compound of Formula (TV).
  • the method is effective at sensitizing tumor cells to a compound of Formula (XV).
  • the method changes (e.g., enhances) the efficacy of a compound of Formula (TV).
  • the tumor cells are resistant to treatment with a compound of Formula (IV) alone.
  • the compound of Formula (IV) can be, for example, the compound No. 12 havin the structure of;
  • the immune checkpoint inhibitor can be, for example, a peptidomimetie compound that inhibits the immunosuppressive signal induced by an immune checkpoint, e.g. PI , FD-L1, PD-L2, CTL-4 and/or VISTA.
  • an immune checkpoint e.g. PI , FD-L1, PD-L2, CTL-4 and/or VISTA.
  • Non-limiting exemplary psptidomimetic compounds are disclosed in U.S. Patent Nos. 8,907,033; 9,044,442;
  • the small molecule immune checkpoint inhibitor can be, for example, CA-170 (previously AUPM 170), a first-in-class oral, small molecule antagonist that selectively targets PD-Ll, PD-L2 and V-domain ig suppressor of T cell activation (VISTA) immune checkpoints
  • CA- 170 is currently being evaluated in a phase I trial for the treatment of advanced solid tumors or lymphomas (see Abstract 4861, AACR 107th Annual Meeting 2016; April 16-20, 2016; New La, LA; developed by Cutis and Aurigene).
  • the small molecule immune checkpoint inhibitor can be, for example, one or more of the Bristol-Myers Squibb (BMS) compounds based on the (2-methyl- 3-biphenylyl) methanol scaffold disclosed in WO2015034820, the content of which is hereby incorporated by reference herein in its entirety.
  • BMS Bristol-Myers Squibb
  • he small molecule immune checkpoint inhibitors can be, for example, compounds 8, 37, 202 and 242 of WO2015034820 (designated herein as BMS-8, BMS-37, BMS-202 and BMS-242) and having the structure of:
  • BMS-202 and BMS-242 bind directly to PD-Li and not PD-1 and effectively dissociate a preformed PD-1/PD-L1 complex in vitro. NMR studies indicate these molecules block PD-1/PD-L1 interaction by inducing PD-LI dimerization through PD-1 interacting surface.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA. as defined herein, combined with an effective amount of BMS-202.
  • the method is effective at sensitizing tumor cells to BMS-202.
  • the method changes (e.g., enhances) the efficacy of BMS-202.
  • the tumor cells are resistant to treatment with BMS-202 alone.
  • the small molecule immune checkpoint inhibitor can be, for example, an immune checkpoint-specific peptide aptamer.
  • the small molecule immune checkpoint inhibitor can be, for example, an immune checkpoint-specific affimer.
  • Affimers are peptide aptamers that are engineered into a modified human protease inhibitor Stefin A scaffold (see, for example. U.S. Patent No. 9,447,170, the content of which is hereby incorporated by reference herein in its entirety for any purpose).
  • the small molecule immune checkpoint inliibitor may comprise an amino acid sequence having at least 80% identity to the Stefin A scaffold polypeptide sequence of SEQ ID NO. : 987. .
  • the small molecule immune checkpoint inhibitor nsay comprise an amino acid sequence having at least 80% identity to the modified Stefin A scaffold polypeptide sequence of SEQ ID NO, : 988.
  • the small molecule immune checkpoint inhibitor can be, for example, a PD-Ll-specific affimer (e.g. PDL1-141 Or PDL1- 179; see, for example, Avacta Life Sciences poster entitled "Generation and Formatting of Mfmier® Biotherapeutics for the Inhibition of the PD-Ll/PD-1 Pathway" 14 th Annual Discovery on Target, September 19-22, 2016, Boston, MA).
  • a PD-Ll-specific affimer e.g. PDL1-141 Or PDL1- 179; see, for example, Avacta Life Sciences poster entitled "Generation and Formatting of Mfmier® Biotherapeutics for the Inhibition of the PD-Ll/PD-1 Pathway" 14 th Annual Discovery on Target, September 19-22, 2016, Boston, MA.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount, of a PD-L1 -specific affimer.
  • he method is effective at sensitizing tumor cells to PD-Ll-specific affimer.
  • the method changes (e.g., enhances) the efficacy of PD-Ll- specific affimer.
  • the tumor cells are resistant to treatment with PD- Ll-specific affimer alone.
  • the PD-Ll-specific affimer can comprise PDL1- 141 and/or PDL1-179.
  • the small molecule immune checkpoint inhibitor can be, for example, a bromodomain and extraterminal domain (BET) inhibitor e.g.. JQ 1 (also known as TEN-01Q in clinical trials NCT02308761 and NCT01987362).
  • a bromodomain and extraterminal domain (BET) inhibitor can be an inhibitor of BRD4.
  • the present disclosure provides a method for enhancing an immune response against a tumor compri sing administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of a BET inhibitor, in certain embodiments, the method is effective at sensitizing tumor cells to BET inhibitor.
  • the method changes (e.g., enhances) the efficacy of BET inhibitor, hi certain embodiments, the tumor cells are resistant to treatment with BET inhibitor alone.
  • the BET inhibitor can comprise an inhibitor of BRD4, e.g. JQ1.
  • the immune checkpoint inhibitor is a monoclonal or polyclonal antibody directed at PD-1 , PD-1 .
  • Exemplaiy PD- 1 immune checkpomt inhibitors that may be combined with a modulator of KRAS signaling, as disclosed herein, include, but are not limited, to:
  • OpdivoTM developed by Bristol-Myers Squibb, Medarex
  • Nivolumab is a fully human immunoglobulin (Ig) G4 monoclonal antibody directed against the negative immuno-regulatory human cell surface receptor programmed death- 1 (PD- 1, PCD-1).
  • Nivolumab can bind to and block the activation of PD- 1 by its ligands programmed cell death ligand 1 (PD-L1), overexpressed on certain cancer cells, and programmed cell death ligand 2 (PD-L2). which is primarily expressed on APCs. This can result in the activation of T-cells and cell-mediated immune responses against tumor cells or pathogens.
  • Compositions and methods of using and making nivolumab are disclosed, for example, in U.S. Patent Nos. 9,387,247; 8,779, 105; 8,779,105; 8, 168, 179; and 8,008,449, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a RAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of nivolumab.
  • the method is effective at sensitizing tumor cells to nivolomab.
  • the method changes (e.g., enhances) the efficacy of nivolomab.
  • the tumor cells are resistant, to treatment with nivolomab alone,
  • immunoglobulin G4 anti -(human programmed cell death 1 )
  • Pembrolizumab is a humanized monoclonal immunoglobulin (Ig) G4 antibody directed against human cell surface receptor PD-1 (programmed death- 1 or programmed cell death- 1). Upon administration, pembrolizumab can bind to PD-1, an inhibitory signaling receptor expressed on the surface of activated T ceils, and block the binding to and activation of PD-1 by its ligands, which can result in the activation of T-cell-mediated immune responses against tumor ceils.
  • Compositions and methods of using and making pembrolizumab are disclosed, for example, in U.S. Patent Nos. 8,354,509; 8,900,587; 8,952, 136; and 9,220,776, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA as defined herein, combined with an effective amount of pembrolizumab.
  • the method is effective at. sensitizing tumor cells to pembrolizumab.
  • the method changes (e.g., enhances) the efficacy of pembrolizumab.
  • the tumor cells are resistant to treatment with pembrolizumab alone.
  • Immunoglobulin G4 anti-(human programmed cell. death protein 1)
  • compositions and methods of using and making JS001 are disclosed, for example, in International Patent Application No. PCT/CN2014/072574, the content of which is hereby incorporated by reference herein in its entirety for any purpose
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor in a subject comprising admin stering to the subject an effective amount of a modulator of KRAS signaling, e.g.. a KRAS aiRNA as defined herein, combined with an effective amount of JS001.
  • the method is effective at sensitizing tumor cells lo JS001. in certain embodiments., the method changes (e.g., enhances) the efficacy of JS00 L
  • the tumor cells are resistant to treatment with JS001 alone.
  • Immunoglobulin G4 anti-(human programmed cell death protein 1)
  • REGN2810 is a human monoclonal antibody directed against the negative immunoregulatory human cell surface receptor programmed cell death 1 (PD-I) protein. Upon administration, anti-PD-1 monoclonal antibody REGN2810 can bind to PD-1, inhibit its binding to the PD-1 ligand programmed cell death- 1 iigand 1 (PD-L1), and prevent the activation of its downstream signaling pathways.
  • PD-I human cell surface receptor programmed cell death 1
  • compositions and methods of using and making REGN2810 are disclosed, for example, in the published U.S. Patent Application No. 2015/0203579, the content of which is hereby incorporated by reference herein in its entirety for any purpose
  • the present disclosure provides a method ibr enhancing an immune response against a tumor compri sing administering an effective amount of a modulator of KRAS signaling, e.g.. a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immun response against, a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of REGN2810.
  • the method is effective at sensitizing tumor cells to REGN2810.
  • the method changes (e.g., enhances) the efficacy of REGN2810.
  • the tumor cells are resistant to treatment with REGN2810 a!one.
  • Immunoglobulin G4 ⁇ kappa anti-[Homo sapiens PDCD1 (programmed cell death 1, PD- L CD279)j, humanized monoclonal antibody;
  • gamma4 heavy chain (1-443) [humanized VH (Homo sapiens IGHV3- 7*01 (90.80%) -(IGHD) -IGHJ4*01) [8.8,9] (1-1 16) -IGHG4*01 (CHI (117-214), hinge S 10>P (224) (215-226), CH2 (227-336), CH3 (337- 441), CHS (442- 443)) ( 117-443)], (130-214') disulfide with kappa light chain ( ⁇ -214 5 ) [humanized V-KAPPA (Homo sapiens IGKV1- 39*01 (87.40%) -IGKJ 1 *01) [6.3.9] ( ⁇ -107') -Homo sapiens
  • Solid tumors e.g., breast cancer, lung cancer
  • SHR- 1210 is a monoclonal antibody directed against the negative immunoregulatory human cell surface receptor programmed death- 1 (PD-1). Upon administration, anii-PD-1 monoclonal antibody SHR-1210 can bind to and block the binding of PD- 1 to its iigands programmed cell death ligand 1 (PD-L1), overexpressed on certain cancer cells, and programmed cell death ligand 2 (PD-L2), which is primarily expressed on antigen presenting cells (APCs).
  • PDCs antigen presenting cells
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effeciive amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of SHR- 1210.
  • the method is effective at sensitizing tumor cells to SHR-1210. hi certain embodiments, the method changes (e.g., enhances) the efficacy of SHR-1210.
  • the tumor cells are resistant to treatment with SHR-1210 alone.
  • Immunoglobulin G4 anti -(human programmed cell death 1 ligand protein PDCD1) (human-M s m sc l s monoclonal MEDI0680 ⁇ 4- chain), disulfide with human- iis musculus monoclonal MEDI0680 ⁇ - chain, dimer
  • MED 10680 is a humanized immunoglobulin (Ig) G4 monoclonal antibody directed against the negative immunoregulatory human eel! surface receptor programmed cell death 1 (PD-1. Upon administration, anti-PD- l monoclonal antibody MEDI0680 can bind to and inhibit PD-1 and its downstream signaling pathways.
  • Ig immunoglobulin
  • PD-1 surface receptor programmed cell death 1
  • compositions and methods of using and making MED10680 are disclosed, for example, in U.S. Patent No. 9,205, 148 and the published U.S. Patent Application No. 2016/0130348, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, In. certain embodiments, the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of MED10680. In certain embodiments, the method is effective at sensitizing tumor cells to MED 10680. In certain embodiments, the method changes (e.g., enhances) the efficacy of MED10680. In certain embodiments, the tumor cells are resistant to treatment with ED10680 alone.
  • PDR.001 is a fully humanized monoclonal antibody that binds to PD-1 with high affinity and inhibits the biological activity of PD-1.
  • anti-PD-1 monoclonal antibody PDR001 can bind to PD-1 expressed on activated T-cells and block the interaction with its ligands, programmed cell death 1 ligand 1 (PD-Ll, PD-lLl) and PD-1 ligand 2 (PD- L2. PD-1L2).
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of PDR001.
  • the method is effective at sensitizing tumor cells to PDR001.
  • the method changes (e.g., enhances) the efficacy of PDR001.
  • the tumor cells are resistant to treatment with PDR001 alone.
  • BGB-317 is a monoclonal antibody directed against the negative immunoregulatory human cell surface receptor programmed cell death 1 (PD-1). Upon administration, anti-PD-1 monoclonal antibody BGB-3 17 can bind to PD-1 and inhibit the binding of PD- 1 to the PD- 1 ligands programmed cell death- 1 ligand 1 (PD-L1), and PD- 1 Hgatid 2 (PD-L2).
  • PD-1 negative immunoregulatory human cell surface receptor programmed cell death 1
  • anti-PD-1 monoclonal antibody BGB-3 17 can bind to PD-1 and inhibit the binding of PD- 1 to the PD- 1 ligands programmed cell death- 1 ligand 1 (PD-L1), and PD- 1 Hgatid 2 (PD-L2).
  • compositions and methods of using and making BGB-317 are disclosed, for example, in U.S. Patent Nos. 9,217,034 and 8,735,553, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present, disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA as defined herein, combined with an effective amount of BGB-317.
  • the method is effective at sensitizing tumor cells to BGB-317.
  • the method changes (e.g., enhances) the efficacy of BGB-317.
  • the tumor cells are resistant to treatment with BGB-317 alone.
  • TSR-042 is a humanized monoclonal antibody directed against the negative immunoregulatory human cell surface receptor programmed cell death 1 (PD-1 ; programmed death-1), with potential smmune checkpoint inhibitory d antineoplastic activities, Upon administration, anti-PD- 1 monoclonal antibody TSR-042 can bind to and inhibit PD-1 and its downstream signaling pathways.
  • PD-1 negative immunoregulatory human cell surface receptor programmed cell death 1
  • TSR-042 can bind to and inhibit PD-1 and its downstream signaling pathways.
  • compositions and methods of using and making TSR-042 are disclosed, for example, in U.S. published Patent Application Patent No. 2016/0075783, the content of which is hereby incorporated by reference herein in its entirety for any purpose
  • the present disclosure provides a method for enhancing an immune response against a tumor comprisiiig administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of TSR-042.
  • the method is effective at sensitizing tumor cells to TSR-042.
  • the method changes (e.g., enhances) the efficacy of TSR-042.
  • the tumor cells are resistant to treatment with TSR-042 alone.
  • PF-06801591 is an inhibitor of the human inhibitory receptor programmed cell death 1 (PD-1; PDCDl ), with potential immune checkpoint inhibitory and antineoplastic activities.
  • PD-1 human inhibitory receptor programmed cell death 1
  • anti-PD- 1 checkpoint inhibitor PF-06801591 can target and bind to PD- 1 and blocks the interaction between PD-1 and its ligands, PD- 1 ligand 1 (PD-L1) and PD-1 ligand 2 (PD-L2).
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of PF-06801591.
  • the method is effective at sensitizing tumor cells to PF-06801591.
  • the method changes (e.g.. enhances) the efficacy of PF-06801591.
  • the tumor ceils are resistant to treatment with PF-06801591 alone.
  • HfirinCAR-PD- 1 cells (PD-1 Antibody Expressing CAR-T Cells)
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA. as defined herein, In certain embodiments, the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of NCH anti-PD- 1 CAR. in certain embodiments, the method is effective at sensitizing tumor cells to NCH anti-PD- 1 CAR. In certain embodiments, the method changes (e.g.. enhances) the efficacy of NCH anti-PD- 1 CAR. In certain embodiments, the tumor cells are resistant to treatment with NCH anti-PD-1 CAR alone.
  • compositions and methods of using and making pidilizumab are disclosed, for example, in U. S. Patent Nos. US7332582; US8686119; US8747847; US9309308 and US9416175, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of pidilizumab.
  • the method is effective at sensitizing tumor cells to pidilizumab.
  • the method changes (e.g., enhances) the efficacy of pidilizumab.
  • the tumor cells are resistant to treatment with pidilizumab alone.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, hi certain embodiments, the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of a preclinical inhibitor of PDl shown in TABLE 7.
  • the method is effective at sensitizing tumor cells to the preclinical inhibitor of PDl, In certain embodiments, tiie metliod changes (e.g.. enhances) the efficacy of the preclinical inhibitor of PDl. In certain embodiments, the tumor cells are resistant to treatment with the preclinical inhibitor of PDl alone.
  • Exemplary PD-L1 immune checkpoint inhibitors that may be combined with a modulator of KRAS signaling, as disclosed herein, include, but are not limited, to:
  • Atezolizumab is a human, Fc optimized, monoclonal antibody directed against the protein ligand PD-Ll (programmed cell death- 1 ligand 1), with potential immune checkpoint inhibitory and antineoplastic activities.
  • PD-Ll is overexpressed on many human cancer cell types and on various tumor-infiltrating immune cells
  • Atezolizumab can bind to PD-Ll, blocking its binding to and activation of its receptor programmed death 1 (PD-1) expressed on activated T-eells, which may enhance the T-cell-mediated immune response to neoplasms and reverse T-cell inactivation.
  • PD-1 programmed death 1
  • atezolizumab can also prevent binding of this ligand to B7.
  • Atezolizumab is modified in such a. way that it does not induce either antibody-dependent cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • compositions and methods of using and making atezolizumab are disclosed, for example, in U.S. Patent No. 8,217.149, the content of which is hereby incorporated by reference herein in its entirety for any purpose
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g.. a KRAS aiRNA. as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of atezolizumab.
  • the method is effective at sensitizing tumor cells to atezolizumab.
  • the method changes (e.g., enhances) the efficacy of atezolizumab.
  • the tumor cells are resistant to treatment with atezolizumab alone.
  • Immunoglobulin G l anti-(human CD antigen CD274) (human
  • MSBOG 10718C monoclonal MSB0010718C heavy chain
  • disulfide with human monoclonal MSB0010718C light chain
  • aimer Code .name: MSBOG 10718C, A09-246-2 (developed by Merck Serono and Pfizer)
  • Avelumab is a human immunoglobulin Gl (IgGl) monoclonal antibody directed against the human immunosuppressive ligand programmed death-ligand 1 (PD-Ll) protein, with potential immune checkpoint inhibitory and antineoplastic activities.
  • avelumab can bind to PD-Ll and prevent the interaction of PD-Ll with its receptor programmed cell death protein ⁇ (PD- 1 ). This can inhibit the activation of PD-1 and its downstream signaling pathways. This may restore immune function through the activation of cytotoxic T-lymphocytes (CTLs) targeted to PD-Ll-overexpressing tumor cells.
  • CTLs cytotoxic T-lymphocytes
  • avelumab can induce an antibody-dependent cellular cytotoxic (ADCC) response against PD- Ll -expressing tumor cells.
  • ADCC antibody-dependent cellular cytotoxic
  • compositions and methods of using and making avelumab are disclosed, for example, in the published International Patent Publication Nos. WO2013079174 and WO2016137985, the contents of which are hereby incorporated by reference herein in their enti eties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of avelumab.
  • the method is effective at sensitizing tumor cells to avelumab.
  • the method changes (e. g., enhances) the efficacy of avelumab.
  • the tumor cells are resistant to treatment with avelumab alone. Durvalumab
  • Immunoglobulin GL anti-(human protein B7-H1) human monoclonal
  • Durvalumab is an Fc optimized monoclonal antibody directed against programmed cell death- 1 ligand 1 (PD-Ll; B7 homolog 1; B7H1), with potential immune checkpoint inhibitory and antineoplastic activities.
  • PD-Ll programmed cell death- 1 ligand 1
  • durvalumab can bind to PD-Ll, thereby blocking its binding to and activation of its receptor programmed death 1 (PD-1) expressed on activated T-cells. This may reverse T-cell inactivation and activate the immune system to exert a cytotoxic T-lymphocyte (CTL) response against PD-Ll-expressing tumor cells.
  • CTL cytotoxic T-lymphocyte
  • the Fc region of durvalumab is modified in such a way that it does not induce eitlier antibody-dependent cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • compositions and methods of making and using durvalumab are disclosed, for example, in U.S. Patent No. 8,779, 108 and the published U.S. Patent Application No. 2016/0222120, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA as defined herein, combined with an effective amount of durvalumab.
  • the method is effective at sensitizing tumor cells to durvalumab.
  • the method changes (e.g., enhances) the efficacy of durvalumab.
  • the tumor ceils are resistant to treatment with durvalumab alone.
  • AMP-224 is a recombinant B7-DC- Fc-fusion protein composed of the extracellular domain of the PD- l ligand programmed cell death ligand 2 (PD-L2, B7-DC) and the Fc region of human immunoglobulin (Ig) Gl
  • AMP-224 is a recombinant B7-DC Fc-fusion protein that binds to PD-1.
  • an optimized treatment regimen with AMP-224 eradicated established tumors of the animals and conferred long-term protective anti-cancer immunity,
  • compositions and methods of making and using AMP-224 are disclosed, for example, in the International Publication Nos. WO2010027827 and WO2011066342, the contents of which are hereby incorporated by reference herein in their entireties for any purpose,
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of AMP-224.
  • the method is effective at sensitizing tumor cells to AMP-224.
  • the method changes (e.g., enhances) the efficacy of AMP-224.
  • the tumor cells are resistant to treatment with AMP-224.
  • MDX- 1105 is an antt-PD-L 1 antibody that is in Phase ⁇ / ⁇ clini cal trials for the treatment of advanced cancers, including hematologic malignancies, melanoma, renal cell carcinoma and solid tumors (Brahmer et ah N. Engl. J. Med. (2012) 366:2455-65).
  • compositions and methods of making and using AMP-224 are disclosed, for example, in U.S. Patent Nos. 7,943,743; 8,383,796; 9, 102,725; 9,212,224 and 9,273,135, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprismg administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprismg administering a effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of MDX-1105.
  • the method is effective at sensitizing tumor cells to MDX-1105.
  • the method changes (e.g., enhances) the efficacy of MDX-1105.
  • the tumor cells are resistant to treatment with MDX-1105, LY3300054
  • LY3300054 is an anti-PD-Ll antibody that has entered Phase I clinical trial in June 2016 for the treatment of advanced refractory solid tiunors (Clinical Trial ID; NCT02791334)
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of K AS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of LY3300054.
  • the method is effective at sensitizing tumor cells to LY3300054.
  • the method changes (e.g., enhances) the efficacy of LY33G0054.
  • the tumor cells are resistant to treatment with LY3300054.
  • Alphamab is an anti-PD-Ll heavy chain antibody selected from a large camel naive phage displ y Nanobody library.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of Alphamab.
  • the method is effective at sensitizing tumor cells to Alphamab.
  • the method changes (e.g., enhances) the efficacy of Alphamab.
  • the tumor cells are resistant to treatment with Alphamab.
  • compositions a nd methods of I using and making STI-1014 are I disclosed in US9175082, the j iSTI-1014 Sorrento
  • compositions a nd methods of I using and making Kadmon patent j anti- PD-L1 are disclosed in j
  • compositions a nd methods of using and making R-Pha m patent anti- PD-Ll are disclosed in
  • compositions a nd methods of using and making BeiGene patent anti- PD-Ll are disclosed in
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signal ing, e. g., a KRAS aiRNA, as defined herein, combined with an effective amount of a preclinical inhibitor of PD- Ll shown in TABLE 7.
  • the method is effective at sensitizing tumor cells to the preclinical inhibitor of PD-L l .
  • the method changes (e.
  • PD-L2 immune checkpoint inhibitors that may be combined with a modulator of KRAS signaling, as disclosed herein, include, but are not limited, to: rIiIgM12B7
  • B7-DC cross-linking antibody rHIgM12B7 binds and crosslinks the B7 co-stimulatory family member B7-DC (PD-L2) on dendritic cells (DCs), antigen presenting cells (APCs) that play a crucial role in the human immune response.
  • PD-L2 dendritic cells
  • APCs antigen presenting cells
  • compositions and methods of using and making rHIgM12B7 are disclosed in the published U.S. Patent Application No. 2016/0122431 and U.S. Patent No. 9,255, 147, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, in certain embodiments, the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e. g., a KRAS aiRNA, as defined herein, combined with an effective amount of rHIgM12B7. In certain embodiments, the method is effective at sensitizing tumor cells to rHIgM12B7.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA
  • the method changes (e.g., enhances) the efficacy of rHIgMI2B7.
  • the tumor cells are resistant to treatment with rHIgM12B7 alone Dana-Farber patent anti-PD-L2 (preclinical)
  • CTLA-4 immune checkpoint inhibitors thai may be combined with a modulator of KRAS signaling, as disclosed herein, include, but are not limited, to:
  • immunoglobulin GL anti-(human CTLA-4 (antigen)) (human ⁇ 1 ⁇
  • Ipilimumab is a recombinant human immunoglobulin (Ig) Gl monoclonal antibody directed against the human T-cell receptor cytotoxic T-lymphocyte-associated antigen 4 (CTLA4), with immune checkpoint inhibitory and antineoplast c activities.
  • Ipilimumab ca bind to CTLA4 expressed on T-cells and inhibit the CTLA4 ⁇ mediated downregulation of T- cell activation. This can lead to a cytotoxic T- lymphocyte (CTL)-mediated immune response against cancer cells.
  • CTL cytotoxic T- lymphocyte
  • compositions and methods of making and using i ilimumab are disclosed, for example, in the U. S. Patent Nos. 9,358,289; 9,320,81 1; 8,784,815; 9,062, 111; 8,685,394; 8,475,790; 8,119,129; 8,449,886; 8, 110, 194; 6,984,720 and the International Publication Nos.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amotsnt of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of ipilimumab.
  • the method is effective at sensitizing tumor cells to ipilimumab.
  • the method changes (e.g., enhances) the efficacy of ipjiimumab.
  • the tumor cells are resistant to treatment with ipilimumab alone.
  • Tremelimumah is a human immunoglobulin (Ig) G2 monoclonal antibody directed against the human T-cell receptor protein cytotoxic T-lymphocyte-associated protein 4 (CTLA4), with potential immune checkpoint inhibitory and antineoplastic activities.
  • CTLA4 cytotoxic T-lymphocyte-associated protein 4
  • Tremelimumah ca bind to CTLA4 on activated T-iymphocytes and block the binding of the antigen-presenting cell ligands B7-1 (CD80) and B7-2 (CD86) to CTLA4, resulting in inhibition of CTLA4-mediated downregulation of T-cell activation. This can promote the interaction of B7-1 and B7-2 with another T-cell surface receptor protein CD28, and result in a B7-CD28-mediated T-cell activation that is unopposed by CTLA4-mediated inhibition.
  • Ig human immunoglobulin
  • compositions and methods of making and using tremelimumah are disclosed, for example, in U. S. Patent Nos. 8,883,984; 8,491,895; 8,685,394; 7,824,679; 8, 143,379; 7,411,057; 7, 132,281; 7, 109,003; 6,682,736 and the International Publication Nos. WO2016030455, WO2015173267, WO2011045704, O2007U3648, WO2006101691, WO2006101692, WO2006096491, WO2006048749, WO2005092380, the contents of which are e eb ⁇ ' incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA as defined herein, combined with an effective amount of tremelimumah.
  • the method is effective at sensitizing tumor ceils to tremelimumah.
  • the method changes (e.g., enhances) the efficacy of tremelimumab.
  • the tumor cells are resistant to treatment with tremelimumab alone.
  • AGEN-1884 developed by Agenus is an anti-CTLA ⁇ 4 antibody that is Phase I clinical trials for the treatment of solid tumors (Clinical Trial ID: NCT02694822).
  • the present disclosure providss a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e. g. 5 a KRAS aiRNA, as defined herein, combined with an effective amount of AGEN-1884.
  • the method is effective at sensitizing tumor cells to AGEN-1884.
  • the method changes (e.g., enhances) the efficacy of AGEN-1884.
  • the tumor cells are resistant to treatment with AGEN-1884 alone.
  • Exemplary VISTA immune checkpoint inhibitors thai may be combined with a modulator of KRAS signaling, as disclosed herein, include, but are not limited, to: Janssen patent anti-VISTA antibody (preclinical)
  • compositions and methods of making and using Janssen patent, anti-VISTA antibody are disclosed, for example, in WO2015097536, the content of which is hereby incorporated by reference herein in its entirety.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method foi" enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of Janssen patent anti-VISTA a tibody.
  • the method is effective at sensitizing tumor cells to Janssen patent anti-VISTA antibody.
  • the method changes (e.g., enhances) the efficacy of Janssen patent anti-VISTA antibody.
  • the tumor cells are resistant to treatment with Janssen patent anti-VISTA . ntibody alone.
  • compositions and methods of making and using igenica patent anti-C10orf54 antibody are disclosed, for example, in O20.14197849 and WO2016094837, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of Igenica patent anii-C lOorf antibody.
  • the method is effective at sensitizing tumor cells to Igenica patent anti-C10orf54.
  • the method changes (e.g., enhances) the efficacy of Igenica patent anti-C10orf54 antibody.
  • the tumor cells are resistant, to treatment with Igenica patent anti-ClQorf54 antibody alone.
  • compositions and methods of making and using Amplimmune patent anti-B7-H5 antibody are disclosed, for example, in WO2014190356 and the published U.S. Patent Application No. 20.16/0096891, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, hi certain embodiments, the present disclosure provides a method for enhancing an immune response against a tumor comprising administering an effective amount of a modulator of KRAS signaling, e.g., a KRAS aiRNA, as defined herein, combined with an effective amount of Igenica patent Amplimmune patent anti-B7-H5 antibody.
  • the method is effective at sensitizing tumor cells ' to Ampl immune patent anti-B7-H5 antibody, m certain embodiments, the method changes (e.g., enhances) the efficacy of Amplimmune patent anti-B7-H5 antibody. In certain embodiments, the tumor cells are resistant to treatment with Amplimmune patent anti-B7-H5 antibody alone.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a co-administered therapeutic agent.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA
  • the change in the efficacy of a therapeutic agent, as a result of the co- dministration of a modulator of KRAS signaling as defined herein can be evaluated in subcutaneous tumor animal models at endpoints such as the percent test/control (%T/C) tumor weights calculated on each day that tumors are measured, tumor growth delay, net log cell kill, median days to a defined tumor weight or to a specified number of tumor doublings, and tumor regression.
  • %T/C percent test/control
  • the lowest calculated %T/C seen over time can be defined as the optimal %T/C because it defines the greatest level of activity seen with the therapeutic agent.
  • the rate and duration of partial and complete tumor regressions can also be considered to be clinically relevant endpoints.
  • a T/C 0% means no tumor growth.
  • a T/C 100% means no antitumor activity, i.e., the treated and control tumors grew equally.
  • a T/C equal to or less than 42% is considered significant antitumor activity by the Drag Evaluation Branch of the Division of Cancer Treatment (NCI).
  • a T/C value ⁇ 10% is considered to indicate highly significant antitumor activity, and is the level used by NCI to justify a clinical trial if toxicity, formulation, and certain other requirements are met (termed DN- 2 level activity).
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KR AS aiRNA, wherein the modulator of KRAS signaling changes (e.g.. enhances) the efficacy of radiotherapy,
  • a modulator of KRAS signaling e.g., a KRAS aiRNA as defined herein, can be combined with radiotherapy.
  • the method is effective at enhancing the efficacy of the radiotherapy.
  • the present disclosure provides a method of treating cancer comprising admiriistering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of an anti-angiogenesis agent.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with an anti-angiogenesis agent.
  • the method is effective at enhancing the efficacy of the anti-angiogenesis agent.
  • Non-limiting examples of anti-angiogenesis agents include, for example, MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (inatrix- etalloproteinase 9) inhibitors, and COX- 11 (eyelooxygenase 11.) inhibitors, rapamycin, temsirolimus (CCI-779). everoiiraus (RADOOl), sorafenib, sxinitinib, and bevacizumab.
  • COX- ⁇ inhibitors include but are not limited to, CelebrexTM (alecoxib), valdecoxib, and rofecoxib.
  • Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172, WO 96/27583, WO 98/07697, WO 98/035 16, WO 98/34918, WO 98/34915, WO 98/33768, WO 98/30566, WO 90/05719, WO 99/52910, WO 99/52889, WO 99/29667, PCX International Application No. PCT IB98/01113, U. S. Patent Nos. 5,863,949 and 5,861,510, all of which are incorporated by reference herein in their entireties for any purpose.
  • MMP inhibitors include, but are not limited to, AG-3340, RO 32-3555, and RS 13-0830.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a chemotherapeutic agent.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with a chemotherapeutic agent.
  • the chemotherapeutic agent includes, but not limited to, antimetabolites, antibiotics, alkylating agents, plant alkaloids, and/or hormonal agents.
  • the method is effective at enhancing the efficacy of the chemotherapeutic agent.
  • ehemotherapeulie agents e.g., suitable for use in compositions and methods described herein
  • alkaloids including isiicrotubule inhibitors (e.g., vincristine, vinblastine, and vindesine, etc.), microtubule stabilizers (e.g., paclitaxel (Taxol), and docetaxel (taxotsre), etc.), and chromatin function inhibitors, including topoisomerase inhibitors, such as epipodophyllotoxins (e.g., etopossde ' (VP- 16), and teniposide (VM-26), etc.), and agents that target topoisomerase I (e.g., camptothecin and isirinotecan (CPT-11), etc.);
  • isiicrotubule inhibitors e.g., vincristine, vinblastine, and vindesine, etc.
  • microtubule stabilizers e.g., paclitaxel (Taxol), and docetaxel (taxotsre), etc.
  • chromatin function inhibitors including
  • covalent DN A- binding agents (alkylatmg agents), inchiding nitrogen mustards (e.g., mechlorethamine, chlorambucil, cyclophosphamide, ifosphami.de, and busulfan (Myleran). etc.), nitrosoureas (e.g., carmustine, lomustine, and semustine, etc.), and other alkylating agents (e.g., dacarbazine, hydroxymethylmelamine, thiotepa, and mitomycin, etc.);
  • nitrogen mustards e.g., mechlorethamine, chlorambucil, cyclophosphamide, ifosphami.de, and busulfan (Myleran). etc.
  • nitrosoureas e.g., carmustine, lomustine, and semustine, etc.
  • other alkylating agents e.g., dacarbazine, hydroxymethylmelamine,
  • noncovalent DNA-binding agents including nucleic acid inhibitors (e.g., dactinomycin (actinomycin D), etc.), anthracyclines (e.g., daunorubic n (daunomycin, and cerubidine), doxorubicin (adriamycin), and idarubicin (idamycin), etc.), anthracenediones (e.g., anthracycline analogues, such as mitoxantrone, etc.), bleomycins (Blenoxane), etc., and piicamycin (mithramycin), etc.;
  • nucleic acid inhibitors e.g., dactinomycin (actinomycin D), etc.
  • anthracyclines e.g., daunorubic n (daunomycin, and cerubidine
  • doxorubicin doxorubicin
  • idarubicin idamycin
  • antimetabolites including antifolaies (e.g., methotrexate, Folex, and Mexate, etc.), purine antimetabolites (e.g., 6-mercaptopurme (6-MP, Purinethol), 6-thioguanine (6 ⁇ TG), azathioprine, acyclovir, ganciclovir, chlorodeoxyadenosine, 2- chlorodeoxyadenosine (CdA), and 2'-deoxycoformycin (pentostatin), etc.), pyrimidine antagonists (e.g., fiuoropyrimidinea (e.g., S-fluorouracil (Adracil), 5- iluorodeox uridine (FdUrd) (floxuridine)) etc.), and cylosine arabinosides (e.g., Cytosar (ara-C) and fludarabine, etc.); 5) enzymes, including L-asparaginase, and hydroxyure
  • hormones including glucocorticoids, antiestrogens (e.g., tamoxifen, etc.), nonsteroidal antiandrogens (e.g., fmtamide, etc.), and aromatase inhibitors (e.g., anastrozole (Arimidex), etc.);
  • antiestrogens e.g., tamoxifen, etc.
  • nonsteroidal antiandrogens e.g., fmtamide, etc.
  • aromatase inhibitors e.g., anastrozole (Arimidex), etc.
  • platinum compounds e.g., cisplatin and carboplatin, etc.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of RAS signaling changes (e.g., enhances) the efficacy of a cancer sternness inhibitor.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with, for example, a cancer sternness inhibitor.
  • cancer sternness inhibitor means a molecule that can target, reduce, inhibit, interfere with, or modulate at least one of a plurality of pathways involved in cancer sternness or the expression (e.g.. the production of a functional product, e.g., a protein) of at least one of a plurality of cancer sternness genes,
  • a cancer sternness inhibitor can be, for example, a small molecule that selectively binds a protein encoded by a cancer sternness gene.
  • a cancer sternness inhibitor is a biologic, e.g., a recombinant binding protein or peptide (e.g. APTSTAT3) or nucleic acid (e.g. STAT3 aiRNA see U.S. Patent No, 9,328,345, the content of which is incorporated by reference herein in its entirety for any purpose), or conjugate thereof.
  • a cancel- sternness inhibitor is a cell.
  • a cancer sternness inhibitor is a STAT3 inhibitor that binds to and inhibits a biological activity of STAT3,
  • STAT3 refers to mammalian STATS, In certain embodiments.
  • STATS refers to the human "Signal Transducer and Activator of Transcription 3" having a canonical 770 amino acid sequence (Accession No: P40763-1 ; MP 644805. 1).
  • a modulator of KRAS signaling e.g., a KRAS aiRNA as defined herein
  • a cancer sternness inhibitor can be, for example, a compound chosen from 2-(l- hydroxyethyl)-naphtho[2,3 ⁇ b]fui'an-4,9-dione, 2-acetyl-7-chloro-naphtho[2,3-b]furan-4,9- dione, 2-aeelyl-7-fluoro-naphlho[2,3-b]furan-4,9-dione 5 2-acetylnaphtho[2.3-b]furan-4,9- dionc, or 2-ethyI-naphtho[2,3-b]furan-4,9-dione, prodrugs thereof, derivatives thereof. pharmaceutically acceptable salts of any of the foregoing
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a cancer sternness gene inhibitor chosen from compounds having formula A, shown below,
  • the compound having formula A may also be known as 2- cetylnaphtho[2,3-b]furan- 4,9-dione, napabucasin, or BBI608 and include tautomers thereof.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a K AS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of 2-ac «tylnaphtrio[2 : ,3 ⁇ b]fura3i--4,9-'dione.
  • Non-limiting examples of prodrugs of compounds having formula A include, for example, the phosphoric esters and phosphoric dicstcrs described in U.S. pre-grant Publication No. 2012/0252763 as compound numbers 4011 and 4012 and also suitable compounds described in U.S. Patent No. 9, 150,530.
  • Non-limiting examples of derivatives of compounds having formula A include, for example, the derivatives disclosed in U.S. Patent No. 8,977,803.
  • the disclosures of U.S. pre-grant Publication No. 2012/0252763 and U.S. Patent Nos. 9, 150,530 and 8,977,803 are incorporated by reference herein in their entireties for any purpose.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with, for example, atovaquone, a hydroxy naphthoquinone having the structure of
  • Atovaquone can downregulate cell-surface expression of glycoprotein 130 (gpl30), which is required for the activation of the cancer sternness gene, STAT3 (see, for example, WO 2015050844 Al, the content of which is hereby incorporated by reference in its entirety for any purpose).
  • glycoprotein 130 gpl30
  • STAT3 the cancer sternness gene
  • a modulator of KRAS signaling e.g., a KRAS aiRNA as defined herein
  • a modulator of KRAS signaling can be combined with an RNA interfering agent that targets a cancer sternness gene, e.g. STATS aiRNA
  • an RNA interfering agent that targets a cancer sternness gene e.g. STATS aiRNA
  • U.S. Patent No. 9,328,345 provides exemplary asymmetric interfering RNA duplexes (aiRNA) and uses thereof to silence STATS expression and treat cancer.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a STATS aiRNA.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA
  • the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a STATS aiRNA.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a modulator of KRAS signaling can be combined with 2-acetylnaphtho[2,3-b]furan-4,9-dione thai targets the cancer sternness gene, e.g. STATS.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • atovaquone that downregulates cell-surface expression of glycoprotein 130 (gpl30), which is required for the activation of the cancer sternness gene, STATS.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS' signaling, e.g., a KRAS aiRNA wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of atovaquone.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA as defined herein
  • a modulator of KRAS signaling can be combined with, for example, an inhibitor of a cancer stem cell pathway kinase (CSCPK) such as for example, STK33, MELK, AXL, p70S6K, and PDGFRo.
  • an inhibitor of a cancer stem cell pathway kinase (CSCPK) can be, for example, an inhibitor, and derivatives thereof, that inhibit STK33, disclosed in U.S. Patent No. 9, 187,434, which is hereby enclosed herein by its entirety for any purpose
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with, for example, an aiRNA that targets one or more cancer stem cell pathway kinases (CSCPK).
  • CSCPK cancer stem cell pathway kinases
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a CSCPK.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a cancer stem cell pathway kinase aiRNA that targets STK33.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a modulator of KRAS signaling can be combined with, for example, a cancer stem cell pathway kinase aiRNA that targets MELK.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a cancer stem cell pathway kinase aiRNA that targets MELK.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a modulator of KRAS signaling can be combined with, for example, a cancer stem cell pathway kinase aiRNA that targets AXL.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a cancer stem cell pathway kinase aiRNA that targets AXL.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with, for example, a cancer stem cell pathway kinase aiRNA thai targets p70S6K.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a cancer stem cell pathway kinase aiRNA that targets p70S6K.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein, can be combined with, for example, a cancer stem cell pathway kinase aiRNA that targets PDGFR.
  • the present disclosure provides a method of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a cancer stem cell pathway kinase aiRNA that targets PDGFR.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a cancer stem cell pathway kinase aiRNA that, targets NANOG
  • the present disclosure provides a metliod of treating cancer comprising administering a modulator of KRAS signaling, e.g., a KRAS aiRNA, wherein the modulator of KRAS signaling changes (e.g., enhances) the efficacy of a cancer stem cell pathway kinase aiRNA that targets NANOG.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a cancer stem cell pathway kinase inhibitor including, but not limited to, compounds, or derivatives thereof, disclosed in U.S. Patent No. 8,299, 106 and PCT Patent Application Publication No. WO2014160401.
  • WO2014160401 discloses the disclosures of U.S. Patent No. 8,299, 106 and PCT Patent Application Publication No. WO2014160401 are incorporated by reference herein by reference in their entireties for any purpose.
  • a modulator of KRAS signaling e.g., a KRAS aiRNA, as defined herein
  • a cancer stem cell pathway kinase inhibitor chosen from compounds having formula B, shown below
  • the present disclosure reports on the surprising discovery that a treatment combination of at least one modulator of K AS signaling and a therapeutic agent have a greater effect in inhibiting cancer cells than the added effects of both the at least one modulator of KRAS signaling and the at least one therapeutic agent alone,
  • the present disclosure reports on the surprising discovery that a treatment combination of at least one modulator of KRAS signaling and an immune checkpoint inhibitor have a greater effect, in inhibiting cancer cells than the added effects of both the at least one modulator of KRAS signaling and. the at least one immune ' checkpoint inhibitor alone.
  • the present disclosure reports on the surprising discovery that a treatment combination of at least one modulator of KRAS signaling and an immunotlierapeutic agent have a greater effect in inhibiting cancer cells than the added effects of ' both the at least one modulator of KRAS signaling and the at least, one immunotlierapeutic agent alone.
  • the present disclosure reports on the surprising discovery that a treatment combination of at least one modulator of KRAS signaling and at least one cancer sternness inhibitor have a greater effect in inhibiting cancer cells than the added effects of both the at least one modulator of KRAS signaling and the at least one cancer sternness inhibitor alone.
  • the present disclosure reports on the surprising discovery that, a treatment combination of at least one modulator of KRAS signaling and at least one cancer stem cell pathway kinase inhibitor have a greater effect in inhibiting cancer cells than the added effects of both the at least one modulator of KRAS signaling arid the at least one cancer stem cell pathway kinase inhibitor alone.
  • compositions disclosed herein may be in the form of a pharmaceutical composition.
  • the present disclosure further provides for a pharmaceutical composition comprising an RNA interfering agent, e.g. an aiRNA.
  • the pharmaceutical comprises (as an active agent) at least one RNA interfering agent, e.g., an asymmetrical duplex RNA molecule.
  • the pharmaceutical compositions may comprise a modulator of KRAS signaling, e.g. a KRAS aiRNA, and at least one immunotlierapeutic agent.
  • the pharmaceutical compositions may comprise a modulator of KRAS signaling, e.g. a KRAS aiRNA, and at least one immune checkpoint inhibitor.
  • the pharmaceutical compositions may comprise a modulator of KRAS signaling, e.g. a KRAS aiRNA, and at least one cancer sternness inhibitor, In certain embodiments, the pharmaceutical compositions may comprise a modulator of KRAS signaling, e.g. a KRAS aiRNA, and at least one cancer stem cell pathway kinase (CSCPK) inhibitor.
  • a modulator of KRAS signaling e.g. a KRAS aiRNA
  • CSCPK cancer stem cell pathway kinase
  • the pharmaceutical compositions may comprise a modulator of KRAS signaling, e.g. a KRAS aiRNA, and one or more compounds and at least one pharmaceutically acceptable carrier, where the one or mors compounds are capable of being converted into the at least one therapeutic agent, for example, a cancer sternness inhibitor in a subject, a cancer stem cell pathway kinase (CSCPK) in a subject or an immunotherapeutic agent, in a subject (i.e., a prodrug).
  • a modulator of KRAS signaling e.g. a KRAS aiRNA
  • the one or mors compounds are capable of being converted into the at least one therapeutic agent, for example, a cancer sternness inhibitor in a subject, a cancer stem cell pathway kinase (CSCPK) in a subject or an immunotherapeutic agent, in a subject (i.e., a prodrug).
  • CSCPK cancer stem cell pathway kinase
  • the pharmaceutical composition comprises one or mors carriers selected from the group consisting of a pharmaceutical carrier, a positive-charge carrier, a liposome, a protein carrier, a polymer, a nanopartid , a nanoemulsion, a lipid, and a lipoid
  • the composition may comprise more than RNA interfering agent species for therapeutic applications.
  • the RNA interfeiing agent species may target different genes required for cancer initiation and progression chosen from, for example, a modulator of KRAS signaling as disclosed herein, in combination with RNA interfering agent(s) that target one or more cancer sternness genes and/or cancer stem cell kinase as disclosed herein.
  • the pharmaceutical compositions and formulations of the present disclosure can be the same or similar to the pharmaceutical compositions and formulations developed for siRNA, miRNA, and antisense RNA, except for the RNA ingredient.
  • the siRNA, mi R A, and antisense RN A in the pharmaceutical compositions and formulations can be replaced by the aiRNA molecules of the present disclosure.
  • the pharmaceutical compositions and formulations can also be further modified to accommodate the RNA interfering agent, e.g. aiRNA, of the present disclosure.
  • a "pharmaceutically acceptable salt” or “salt” of an RNA interfering agent is a product of the disclosed RNA interfering agent that contains an ionic bond, and is typically produced by reacting the disclosed RNA interfering agent with either an acid or a base, suitable for administering to a subject.
  • Pharmaceutically acceptable salt can include, but is not limited to, acid addition salts including hydrochlorides, hydrobromides, phosphates, sulphates, hydrogen sulphates, alkylsulphonates, arylsulphonat.es, acetates, benzoates, citrates, maleates, fumarates, succinates, lactates, and tartrates; alkali metal cations such as Na, K, Li, alkali earth metal salts such as Mg or Ca., or organic amine salts.
  • a "pharmaceutical composition” is a formulation containing the disclosed RNA interfering agent, e.g. aiRNA, in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a .tablet, a single pump on an aerosol inhaler, or a vial.
  • the quantity of active ingredient, (e.g., a formulation of the disclosed duplex RNA molecule or salts thereof) in a unit dose of composition is an effective amount and is varied according to the particular ' treatment involved.
  • active ingredient e.g., a formulation of the disclosed duplex RNA molecule or salts thereof
  • the dosage will also depend on the route of administration.
  • RNA interfering agent e.g. aiRNA
  • routes including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, intranasal, and the like.
  • Dosage forms for the topical, or transdermal administration of a RNA interfering agent, e.g. aiRNA, of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active RNA interfering agent e.g. aiRNA
  • the active RNA interfering agent e.g. aiRNA
  • the present disclosure provides a method of treatment comprising adm nostiiing an effective amount of the pharmaceutical composition to a subject in need thereof.
  • the pharmaceutical composition is administered via a route selected from the group consisting of iv, sc, topical, po, and ip.
  • the effective amount is 1 ng to 1 g per day, 100 ng to 1 g per day. or 1 ug to 1 mg per day.
  • RNA interfering agent e.g. aiRNA
  • pharmaceutically acceptable excipient or carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in "Remington: The Science and Practice of Pharmacy, Twentieth Edition,” Lippincott Williams & Wilkins, Philadelphia, PA, which is incorporated by reference herein by reference.
  • Such carriers or diluents include, but are not limited to, water, saline, Ringer's solutions, dextrose solution, and 5% human serum albumin. Liposomes and nonaqueous vehicles such as fixed oils may also be used.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active RNA interfering agent, e.g. aiRNA, use thereof in the compositions is contemplated.
  • Supplementary active RNA interfering agents, e.g. aiRNAs can also be incorporated into the compositions.
  • RNA interfering agent e.g. aiRNA
  • a suitable dosage form prepared by combining a therapeutically effective amount (e.g., an efficacious level sufficient to achieve the desired therapeutic effect through inhibition of tumor growth, killing of tumor cells, treatment, or prevention of cell proliferative disorders, etc.) of a RNA interfering agent, e.g. aiRNA, of the present disclosure (as an active ingredient) with standard pharmaceutical carriers or diluents according to conventional procedures (i.e., by producing a pharmaceutical composition of the disclosure).
  • a therapeutically effective amount e.g., an efficacious level sufficient to achieve the desired therapeutic effect through inhibition of tumor growth, killing of tumor cells, treatment, or prevention of cell proliferative disorders, etc.
  • standard pharmaceutical carriers or diluents i.e., by producing a pharmaceutical composition of the disclosure.
  • RNA interfering agent e.g. aiRNA
  • a suitable dosage form without standard pharmaceutical carriers or diluents.
  • Pharmaceutically acceptable carriers include solid carriers such as lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like.
  • Exemplary liquid carriers include syrup, peanut oil, olive oil, water and the like.
  • the carrier or diluent may include time-delay material known in the art such as glyceryl monostearate or glyceryl distearate, alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate or the like.
  • Other fillers, excipients, flavorants, and other additives such as are known in the art may also be included in a pharmaceutical composition according to this disclosure.
  • solvate represents an aggregate that comprises one or more molecules of a compound of the present disclosure with one or more molecules of a solvent or solvents.
  • Solvates of the compounds of the present disclosure include, for example, hydrates.
  • compositions containing active RNA interfering agent, e.g. aiRNA, of the present disclosure may he manufactured in a manner that is generally known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • Pharmaceutical compositions may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active duplex RNA molecules into preparations thai can be used pharmaceutically.
  • the appropriate formulation is dependent upon the route of administration chosen.
  • RNA interfering agent e.g. aiRNA
  • pharmaceutical composition of the disclosure can be administered to a subject in many of the well-known methods currently used for ehemoihsrapeutic treatment.
  • a RNA interfering agent, e.g. aiRNA, of the present disclosure rnity be injected directly into tumors, injected into the blood stream or body cavities, taken orally, or applied through the skin with patches.
  • aiRNA nanopartieies may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form' ' refers to a physically discrete unit of nanoparticle appropriate for the patient, to be treated. It will be understood, however, that the total daily usage of the compositions of the present disclosure will be decided by the attending physic ian within the scope of sound medical judgment.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. The animal model is also used to achieve a desirable concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • Therapeutic efficacy and toxicity of nanopartieies can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50 (the dose is therapeutically effective in. 50% of the population) and LD50 (the dose is lethal to 50% of the population).
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50.
  • Pharmaceutical compositions which exhibit large therapeutic indices may be useful in some embodiments.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for human use.
  • compositions suitable for parenteral administration may comprise at least one more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions, emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, baeteriosfats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • normal dosage amounts may vary from about 10 ng/kg up to about 100 mg/kg of mammal body weight or more per day, preferably about 1 mg kg/day to 10 mg/kg/day, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature; see, for example, U.S. Pat. No. 4,657,760; 5,206,344; or 5,225,212, the contents of which are hereby incorporated by reference herein in its entireties for any purpose.
  • a composition described herein includes at least one compound and pharmaceutically acceptable salts and solvates thereof and one or more surfactants, in certain embodiments, the surfactant, is sodium lauryi sulfate (SLS), sodium dodecyl sulfate (SDS), or one or more polyoxylgiycerides.
  • the pol oxylglyeeride can be lauroyl polyoxylgiycerides (sometimes referred, to as GeluckeTM) or iinoleoyl polyoxylgiycerides (sometimes referred to as LabrafilTM). Examples of such compositions are disclosed in PCT Patent. Application No. PCT US2014/033566, the content of which is incorporated by reference herein in its entirety for any purpose.
  • the present disclosure provides further embodiments of suitable pharmaceutical formulations having selected particle size distribution and methods for identifying an optimum particle size distribution, suitable drug regimen, dosage and interval, suitable methods of preparing 2-acetylnaphtho[2,3-6]furan-4,9-dione including their crystalline forms, and further specific suitable cancer sternness inhibitors and kinase inhibitors as described in the co-owned PCT applications published as WO 2009/036099, WO 2009/036101 , WO 201 1/1 16398, WO 201 1/116399, WO 2014/169078, and WO 2009/033033, the contents of which are hereby incorporated by reference herein in their entireties for any purpose.
  • a kit comprises (1) at least one compound chosen from a modulator of KRAS signaling and (2) an immune checkpoint inhibitor, an immunotherapeutic, a chemotherapsutic agent, a cancer sternness inhibitor, a cancer stem cell pathway kinase inhibitor or prodrugs thereof, derivatives thereof, pharmaceutically acceptable salts of any of the foregoing, together with instructions for administration and/or use.
  • EXAMPLE 1 KRAS SILENCING IS SUFFICIENT TO DEPLETE PD-L1
  • aiRNA asymmetric interfering RNA
  • aiRNAs were first synthesized in DMT-on mode. Following completion of the synthesis, the solid support was suspended in 600 ⁇ EtOH/MHkOH solution (prepared by mixing 1 volume of 200 proof ethanol wit 3 volumes of 28% NH4OH) and heated at 55 °C for 2 hours. After primary de-protection, EtOH/NiliOH was evaporated and the RNA oligo was dried to a pellet ⁇ of RNA de-protection solution (NMP/TEA.3HF (3:2)) was added and the solution was heated at 65 °C for 1.5 hours. The reaction was then quenched with 400 ⁇ of 1.5 M ammonium bicarbonate. Purification was performed with Clarity® QSP Cartridges (Phenomenex, USA). The annealing of the resulting duplexes was confirmed on 15% PAGE gel. All sequences are shown in FIG. 1A. The location of each of the sequences targeted by aiKRAS is shown in FIG. IB.
  • the human breast adenocarcinoma ceil line, MDA-MB-231, that is heterozygous for the KRAS GOD mutation, was obtained from ATCC (ATCC® CRM-HTB-26TM) and maintained in DMEM supplemented with 10% (vol/Vol) FBS (Gemini Bio Products, USA) and 1% penicillin/streptomycin (Life Technologies, USA).
  • MDA-MB-231 cells were then transfected with multiple aiRNAs targeting KRAS (aiKRAS# l-#4). Cells were seeded to 60mm plates (2 x 10 5 cells/4 mL/well). aiRNA was transfected by Lipofectamine® RNAiMAX (Thermo Fisher, USA) according to the manufacturer's protocol. aiRNA and RNAiMAX were incubated for 20 mi in serum free ⁇ - ⁇ (Thermo Fisher, USA), then added to the cells along with culture medium, 48 hours after transfection, cells were harvested using Aecutase® cell detachment solution (Sigma Aldrich, USA). Scrambled aiRNA was used as a control. The amount of PD-L1, KRAS and actin protein was then determined by Western blot analysis.
  • the level of PD-Ll mRNA was also down -regulated by KRAS silencing with aiKRAS R s 1-4.
  • KRAS silencing down- regulated PD-Ll expression in a transcript on factor dependent manner.
  • Time course experiments showed that aiRNA mediated KRAS silencing was induced very quickly arid reached maximum activity 8 hours post-transfection whereas PD-Ll was down-regulated only after 32 hours post-transfection (Fig. 2C) suggesting the presence of intermediate steps between KRAS silencing and the subsequent reduction in PD-Ll mRNA levels.
  • KRAS activates various down-stream signaling pathways including the RAF/MEK ERK pathway, the P13K/AKT pathway, and the RalGTP pathway.
  • aiKRAS or aiControl transfected KRAS mutant MDA-MB-231 and KRAS wild-type RKO cells were washed twice with ice cold PBS and lysed in lysis buffer [50 mM Hepes (pH 7.5), 1% Nonidet P-40, 150 mM NaCi, 1 mM EDTA, and 1.x protease inhibitor mixture (EMD Miilipore, USA)].
  • Soluble protein was then probed using a Human Phospho-Kinase Antibody Array according to the manufacturer's instructions (R&D Systems; Catalog # ARY003B).
  • a Human Phospho-Kinase Antibody Array according to the manufacturer's instructions (R&D Systems; Catalog # ARY003B).
  • phosphorylated ERK 1/2 was most prominently down-regulated with KR AS depletion in KRAS mutant MDA-MB-231 cells but not in KRAS wild-type RKO cells (Fig. 3 A),
  • KRAS mutant MDA-MB-231 cells were treated with the MEK-specific inhibitor, U0126. Similar to aiKRAS, U0126 also down-regulated PD-Ll expression in MDA-MB-23 1 cells and inliibited ERK phosphorylation (Fig. 4B). These results indicate the ERK pathway is involved in KRAS dependent PD-Ll expression in KRAS mutant cancer cells.
  • AP-1 Activator protein 1
  • the AP-! transcriptional factor is formed by either the homo-dimerization of a Jun family protein (eJun, JunB, or JunD) or by the more stable hetero-dimerization of a Jun family protein with a Fos family protein (cFOS, FGSB, FRA- 1, or FRA-2).
  • cFOS homo-dimerization of a Jun family protein
  • FRA- 1 Fos family protein
  • ai-KRAS# l was transfected into KRAS mutant cancer cell lines (MDA-MB-231, H358, and H460) and a KRAS wild- type cancer ceil line (RKO).
  • Immunoblotting analysis with a Phospho-FRA-1 and FRA- 1 antibody showed thai KRAS depletion down- regulated the phosphorylated and total forma of FRA- 1 in MDA-MB-231, H358, and H460 cells harboring KRAS mutations but not in KRAS wild-type RKO cells (Fig.5 A).
  • FRA-1 is a KRAS dependent transcriptional factor as reported previously by Casalino et l, (2003).
  • KRAS silencing reduced the endogenous FRA-1 binding activity in KRAS mutant MDA-MB-231 cells but not in KRAS wild-type RKO cells. This result was consistent with the observed down-regulation of FRA-1 expression by KRAS silencing in KRAS mutant cells (Fig. 4A).
  • Intron 1 of the PD-L1 gene located approxim tely 5kb downstream from the transcription start site, contains a conserved candidate enhancer element with putative API binding sites.
  • a chromatin immunoprecipitation (ChIP) quantified PGR assay was performed in the presence of anti-phospho-Fra- 1 or cJun antibody using SimpleChIP® Enzymatic Chromatin IP Kit (Cell signaling Technologies. USA) according to the manufacturer's protocol.
  • aiRNA transfected MDA-MB-231 cells were cross-linked by treatment with 1% formaldehyde in culture medium for IGmin at room temperature.
  • nuclear pellets were prepared by treating the cells with Buffers A and B, The nuclear pellets were then suspended in buffer B and treated with SDS buffer and micrococcal nuclease to digest chromatin for 20 min at 37°C. Cell suspensions were then sonicated to elute nuclear chromosomal content in preparation for immunoprecipitation. 2 ,g each of Phospho-FRA-1 and cJun were incubated with the lysate overnight at 4°C. Immunocomplexes were collected with magnetic protein A beads and eluted after extensive washing. Cross-linkage was reversed by heating the solution at 65°C with NaCl and proteinase K.
  • DNA was purified and used as the template for quantitative PGR to amplif ' the region including the AP-1 binding site.
  • the primer pairs used were 5'- GTC AC ATTTC AAGC AGG ATG ACT A-3 ' (SEQ ID NO. : 985) and 5'- GGAAGGGGAGAGAG TTGGATT-3 ' (SEQ ID NO. : 986). Quantitative PGR. was performed as described in Example 1.
  • both FRA- 1 and cJUN were recruited to candidate PD ⁇ LI enhancer sites in MDA-MB-231 cells. Binding of FRA-1 and cJUN to the PD-LI enhancer decreased with KRAS depletion. In contrast, as with PD-L1 gene expression. FRA-1 and cJUN binding to the PD-LI enhancer was not. altered by KRAS depletion in KRAS wild-type RKO cells (Fig. 5C, right side).
  • aiFra-l#L aiFra- l#2 were transfected into KRAS mutant MDA-MB-231 and KRAS wild-type RKO cells, iramunoblotting and quantitative RT-PCR confirmed that transfection of aiFra-l#l or #2 resulted in depletion of FRA-1 protein and mR A (FIG. 6 A and 6B). Moreover, knockdown of FRA-1 also resulted in a marked down-regulation of PD-LI protein and mRNA expression (Fig. 6A and 6B).
  • STAT3 or NF-KB are also known to promote the expression of PD-LI .mRNA by binding directly to the PD-LI promoter region.
  • KRAS silencing did not alter the expression of either STAT3 or NF-KB (see FIGs. 4C and 4D).
  • Luc-MDA-MB-231 a cell line stably expressing luciferase, was purchased .from Cell Biolabs, lac, USA Luc-MD A-MB-231 cells were grown in Dulb cco's modified Eagle's medium (D EM) supplemented with 10% inactivated fetal calf serum (FCS) and transfected with either control aiRNA or aiKRAS#l as described in Example 1.
  • D EM Dulb cco's modified Eagle's medium
  • FCS inactivated fetal calf serum
  • PD-L1 in the control aiRNA or aiKRAS#l transfected Luc- MDA-MB-231 cells was determined by staining the cells with PE conjugated anti-CD274 (BD Biosciences, USA) in Stain Buffer (BD Biosciences, USA) on ice for 20 min and washed once with Sta n Buffer. CD274 positive population was then detected using flow cytometry (Attune Acoustic Focusing Cytometer, Life Technologies, USA) and analyzed with Flowjo software (Flow] o, LLC, LISA). Flow cytometry analysis confirmed that KRAS silencing down-regulated cell surface PD-L1 expression (Fig, 7B).
  • aiRNA-transfected MDA- MB-231 cells were either pulsed or not pulsed with CMV antigen peptide, and then co-cultured with CMV specific cytotoxic T lymphocytes (CTLs) expanded from HL A typed human PBMC from healthy donors,
  • HLA typed PBMCs from healthy donor were purchased from Cellular Technology, Ltd., USA PBMCs were diluted at 5 x 10 6 cells/mL in culture medium (RPMI-1640 supplemented with 10% inactivated human serum and 50 ⁇ 2-mercaptoethanol) and seeded into 24-well plates (5 x 10 6 cells/mL/well).
  • HLA-A*02:01 CMV pp65 peptide (NLVPMVATV) (IB A Lif ⁇ sciences, Germany) was added to a final concentration of 5 ⁇ on day 0 as well as 23 !U/mL IL-2 (R&D Systems, USA) and 5 ng/mL 3L-15 (R&D Systems, USA).
  • CD8 + T cells were isolated from PBMC using a CD8 + T cell Isolation Kit (Miltenyi Biotec, USA) as described by the manufacturer's protocol.
  • the aiRNA transfected Luc-MDA-MB-23 1 cells were incubated with or without HLA- A*02:01 C V pp65 peptide (NL VPMV AT V) for 2 hours at 37°C in 5% CO2 and then washed twice with PBS, CMV peptide loaded or unloaded Luc-MDA-MB-23 1 cells were plated into 96-well plates (2000 cells/well). CMV specific CD8 + T cells were subsequently added to a 96- well plate with an Effector: Target (E/T) ratio of 50: 1 and incubated for 24 hours.
  • E/T Effector: Target
  • Anti-Human PD-L 1 Ab at a final concentration of was used as a control (Clone: ⁇ 1, Functional grade purified, Affymetrix eBioseienee, USA).
  • Live Luc-MDA-MB-23 ⁇ cells were measured by intracellular luciferase activity with XenoLight D-Luciferin K v sait (PerkinElmer, USA) by GloMax® Discovery (Promega, USA). The percent Lysis was calculated with the formula: (Luminescence of CMV peptide pulsed Luc-MDA-MB-231/ Luminescence of CMV peptide un-puised Luc-MDA-MB-23 1)* 100.
  • cytotoxic activity of antigen specific CTLs were significantly enhanced with aiKRAS transfection (pO.01) compared with scramble aiRNA transfected cells.
  • aiRNA targeting PD- L I was compared to the activity of a commercially available functional grade anti-PD-L l antibody. Both treatments enhanced CTL killing activity against MDA-MB-231 cells (Fig. 7A) to a level that was comparable to the cytotoxic activity seen with KRAS depletion.
  • EXAMPLE S PREPARATION OF KRAS aiRNA NANOP ARTICLES (NPs) aiRNAs were synthesized in DMT-on mode as described in Example 1, For certain applications, selected nucleotides in the aiRNAs were modified with a 2' -OH methyl group.
  • Nanoparticles (NPs) loaded with aiRNA were prepared using the double-emulsion process described in WO2014123935, the content of which is hereby incorporated by reference herein in its entirety.
  • aiRNA was dispersed in Ix RNAse-free buffer to make the desired concentrat n (e.g., about 5 ⁇ /' ⁇ ).
  • a volume of aiRNA e.g., about 50 p.g
  • 100 uJL of 500 mM MgCk was mixed with 100 uJL of 500 mM MgCk.
  • the MgCk-aiRNA solution was added drop-wise to the oil/surfactant solution in vial A to form a well-dispersed emulsion without reverse micro-emulsion.
  • the contents of vials A and B were then mixed and stirred for 30 minutes at room temperature.
  • the contents were transferred into 10 centrifuge tubes ( 1.5 mL) and centrifuged for 30 minutes at 13,000g. The supernatant was discarded and the pellet was washed with absolute ethanol (1 mL-) twice. After the alcohol was removed, the resulting pellet was air-dried for 3-4 hours,
  • a polymer- based shell was coated onto the MgP nanoparticle cores already loaded with aiRNA Specifically, biodegradable polymers, PEG(5k)-Poly-L-Lysine ( lOU), Poly-L- Arginine (SOU) were coated onto the cores at a polymer ratio of 2.5 : 1 (PLL: PLR) & a complex ratio of 2.5: 1 (polymer: aiRNA).
  • the average size of the nanoparticl.es was about 70 nm and the surface charge was about +25 mV.
  • the nanoparticles exhibited good plasma stability, cellular uptake and endosomal escape.
  • the human breast adenocarcinoma cell line, MDA-MB-231 (ATCC® CRM-HTB- 26TM) is cultured in DMEM supplemented with 10% (vol/vol) FBS (Gemini Bio Products, USA) and 1% penicillin/streptomycin (Life Technologies, USA). Approximately, 6 X 10 6 cells /mouse are then inoculated subcutaneously into female athymic nude mice.
  • mice are randomized into the following treatment groups: .
  • Irradiations are performed 7- 10 days after inoculation (when tumors were at least 100 mm 3 ) using a Pantak HF-320 320 kV x ⁇ ray unit, (Gulmay Medical, U.K.).
  • the machine is operated at 300 kV, 9.2 rnA, with filtration fitted in the x-ray beam to give a radiation quality of 2.3 mm Cu half -value layer.
  • Mice are positioned at a distance of 350 mm from the x-ray focus, where the dose rate is 0.80 Gy/min.
  • NP preparations comprising either KRAS aiRNAs or an aiRN A control commences on day 1 of the fractionated radiation therapy cycle and is repeated every other day for 8 days (at a dose of 2.5 mg/kg/NP preparation). The treatment is well tolerated with minimal adverse side effects. The size of the tumors is measured every day. Experimental groups contain at least 5 mice/group and are representative of at least 2 independent experiments.
  • Radiotherapy increases tumor cell PD-L1 expression which peaks at about 72 hours after the last dose of radiation and remains elevated as compared to non-treated control mice.
  • the radiotherapy-mediated local tumor control is improved tlirough the administration of aiKRAS#l-#4 nanoparticle formulation.
  • a synergistic anti-tumor response with a T/C value ⁇ 10% is observed in mice that receive radiotherapy in combination with the aiKRASirl-#4 NP as compared to mice that received radiotherapy in combination with the scrambled control aiRNA formulation.
  • mice are randomized into the following treatment groups:
  • Group 2 Control aiRNA NP + paclitaxel
  • Nanoparticle (NP) preparations comprising either aiKRAS#l-#4 or an aiRNA control are administered by intravenous injection to the xenografted mice of Groups L 3 and 4 starting on day 1.
  • the aiR A injections are repeated every other day for 12 days (i.e., a dose of 2, 5 mg/kg/NP preparation on day L 3, 5, 8, and 10).
  • Paclitaxel (Taxol®) is administered to Groups 2 and 4 by intraperitoneal injection on day 2, 4, 6, 9 and 11 at a dose of 10 mg/kg preparation.
  • mice/group contain 10 mice/group and are representative of at, least 2 independent experiments.

Abstract

L'interaction de PD-L1 exprimée sur la surface de cellules tumorales déclenche une signalisation PD-1 dans des lymphocytes T qui contribue à la résistance de cancers à des thérapies de points de contrôle immunitaires émergents. La présente invention démontre que la signalisation RAF/MEK/ERK/FRA induite par KRAS est requise pour l'expression génique PD-L1 dans des cellules tumorales. L'invention concerne ainsi des compositions pour le traitement du cancer comprenant des modulateurs de la signalisation de KRAS combinés à un agent immunothérapeutique qui restaure la sensibilité de cellules tumorales résistantes à l'agent immunothérapeutique. Par exemple, une combinaison d'ARN interférents asymétriques (ARNai) spécifiques de KRAS et d'un inhibiteur de point de contrôle immunitaire s'est révélée améliorer la cytotoxicité des lymphocytes T spécifiques aux cellules tumorales. L'invention concerne également des méthodes permettant d'augmenter l'efficacité thérapeutique de traitements anticancéreux existants par co-administration d'un modulateur de signalisation KRAS.
PCT/US2017/063116 2016-11-22 2017-11-22 Ciblage d'expression du point de contrôle immunitaire induit par kras WO2018098352A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662425358P 2016-11-22 2016-11-22
US62/425,358 2016-11-22

Publications (2)

Publication Number Publication Date
WO2018098352A2 true WO2018098352A2 (fr) 2018-05-31
WO2018098352A3 WO2018098352A3 (fr) 2018-07-05

Family

ID=60703090

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/063116 WO2018098352A2 (fr) 2016-11-22 2017-11-22 Ciblage d'expression du point de contrôle immunitaire induit par kras

Country Status (1)

Country Link
WO (1) WO2018098352A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020014650A1 (fr) * 2018-07-13 2020-01-16 Memorial Sloan Kettering Cancer Center Méthodes d'utilisation d'inhibiteurs pharmacologiques de la signalisation des cytokines de type 2 pour traiter ou prévenir le cancer du pancréas
WO2020081905A1 (fr) * 2018-10-18 2020-04-23 Jounce Therapeutics, Inc Méthodes de traitement du cancer
WO2020205473A1 (fr) * 2019-03-29 2020-10-08 Decerna Pharmaceuticals, Inc. Compositions et procédés pour le traitement de maladies et troubles associés au kras
WO2021016228A1 (fr) * 2019-07-22 2021-01-28 H. Lee Moffitt Cancer Center And Research Institute Inc. Polythérapie pour le traitement de cancers à mutant ras
WO2021061749A1 (fr) * 2019-09-24 2021-04-01 Mirati Therapeutics, Inc. Polythérapies
WO2023087012A1 (fr) * 2021-11-15 2023-05-19 The Board Of Trustees Of The University Of Illinois Méthode de traitement d'un cancer lié à une mutation ras
WO2023172940A1 (fr) * 2022-03-08 2023-09-14 Revolution Medicines, Inc. Méthodes de traitement du cancer du poumon réfractaire immunitaire
US11840533B2 (en) 2017-12-01 2023-12-12 The Board Of Trustees Of The University Of Illinois Pyridinone-based epigenetic modifiers and uses thereof

Citations (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4657760A (en) 1979-03-20 1987-04-14 Ortho Pharmaceutical Corporation Methods and compositions using monoclonal antibody to human T cells
US4820631A (en) 1986-07-30 1989-04-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Deletion mutants and monoclonal antibodies against ras proteins
US4898932A (en) 1985-01-29 1990-02-06 E. I. Du Pont De Nemours And Company Monoclonal antibodies reactive with activated and oncogenic ras p21 proteins
WO1990005719A1 (fr) 1988-11-23 1990-05-31 British Bio-Technology Limited Inhibiteurs de collagenase a base d'acide hydroxamique
US5028527A (en) 1988-02-22 1991-07-02 Applied Bio Technology Monoclonal antibodies against activated ras proteins with amino acid mutations at position 13 of the protein
US5081230A (en) 1987-07-08 1992-01-14 E. I. Dupont Denemours And Company Monoclonal antibodies reactive with normal and oncogenic forms of the ras p21 protein
US5084380A (en) 1985-01-29 1992-01-28 Applied Biotechnology Monoclonal antibodies reactive with activated and oncogenic ras p21 proteins
US5112737A (en) 1988-02-22 1992-05-12 Applied Biotechnology, Inc. Monoclonal antibodies against activated ras proteins with amino acid mutations at position 13 of the protein
US5206344A (en) 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
US5225347A (en) 1989-09-25 1993-07-06 Innovir Laboratories, Inc. Therapeutic ribozyme compositions and expression vectors
US5225212A (en) 1989-10-20 1993-07-06 Liposome Technology, Inc. Microreservoir liposome composition and method
WO1996027583A1 (fr) 1995-03-08 1996-09-12 Pfizer Inc. Derives de l'acide arylsulfonylamino hydroxamique
WO1996033172A1 (fr) 1995-04-20 1996-10-24 Pfizer Inc. Derives d'acide hydroxamique arylsufonyle en tant qu'inhibiteurs de mmp et de tnf
US5693773A (en) 1995-06-07 1997-12-02 Hybridon Incorporated Triplex-forming antisense oligonucleotides having abasic linkers targeting nucleic acids comprising mixed sequences of purines and pyrimidines
WO1998003516A1 (fr) 1996-07-18 1998-01-29 Pfizer Inc. Composes a base de phosphinate inhibiteurs des metalloproteases matricielles
WO1998007697A1 (fr) 1996-08-23 1998-02-26 Pfizer Inc. Derives de l'acide arylsulfonylamino hydroxamique
WO1998030566A1 (fr) 1997-01-06 1998-07-16 Pfizer Inc. Derives de sulfone cyclique
WO1998033768A1 (fr) 1997-02-03 1998-08-06 Pfizer Products Inc. Derives d'acide arylsulfonylaminohydroxamique
WO1998034918A1 (fr) 1997-02-11 1998-08-13 Pfizer Inc. Derives de l'acide arylsulfonylhydroxamique
WO1998034915A1 (fr) 1997-02-07 1998-08-13 Pfizer Inc. Derives du n-hxdroxy-beta-sulfonyl-propionamide et leur utilisation comme inhibiteurs des metalloproteases matrices
WO1999029667A1 (fr) 1997-12-05 1999-06-17 Pfizer Limited Derives d'acide hydroxamique utilises comme inhibiteurs de metalloproteases matricielles
US5965539A (en) 1993-05-18 1999-10-12 Univeristy Of Pittsburgh Inhibitors of prenyl transferases
WO1999052889A1 (fr) 1998-04-10 1999-10-21 Pfizer Products Inc. Hydroxamides de l'acide (4-arylsulfonylamino)-tetrahydropyrane-4-carboxylique
WO1999052910A1 (fr) 1998-04-10 1999-10-21 Pfizer Products Inc. Derives bicycliques de l'acide hydroxamique
US6107094A (en) 1996-06-06 2000-08-22 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US6124133A (en) 1999-10-15 2000-09-26 Isis Pharmaceuticals Inc. Antisense inhibition of fra-1 expression
WO2001014424A2 (fr) 1999-08-24 2001-03-01 Medarex, Inc. Anticorps contre l'antigene ctla-4 humain et utilisation
WO2003086459A1 (fr) 2002-04-12 2003-10-23 Medarex, Inc. Methodes de traitement dans lesquelles sont utilises des anticorps du ctla-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
WO2005092380A2 (fr) 2004-03-26 2005-10-06 Pfizer Products Inc Utilisations d'anticorps anti-ctla-4
WO2006048749A1 (fr) 2004-11-04 2006-05-11 Pfizer Products Inc. Anticorps ctla-4 et inhibiteur de l'aromatase ou traitement combine du cancer du sein
US7056704B2 (en) 2000-12-01 2006-06-06 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
WO2006096491A2 (fr) 2005-03-08 2006-09-14 Pharmacia & Upjohn Company Llc Compositions d'anticorps anti-ctla-4
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2006101691A1 (fr) 2005-03-23 2006-09-28 Pfizer Products Inc. Traitement du cancer de la prostate au moyen d'anticorps anti-ctla4 et d'une hormonotherapie
WO2006101692A1 (fr) 2005-03-23 2006-09-28 Pfizer Products Inc. Association d'anticorps anti-ctla4 et d'indolinone dans le traitement du cancer
WO2007056539A2 (fr) 2005-11-08 2007-05-18 Medarex, Inc. Prevention et traitement de l'enterocolite associee a un traitement par anticorps anti-ctla-4
WO2007067959A2 (fr) 2005-12-07 2007-06-14 Medarex, Inc. Regimes d'escalade de dose d'anticorps ctla-4
WO2007113648A2 (fr) 2006-04-05 2007-10-11 Pfizer Products Inc. Polythérapie à base d'un anticorps anti-ctla4
US7332582B2 (en) 2002-05-23 2008-02-19 Curetech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
WO2008109142A2 (fr) 2007-03-07 2008-09-12 Cold Spring Harbor Laboratory ARNpi ET UTILISATIONS CORRESPONDANTES
WO2009029690A1 (fr) 2007-08-27 2009-03-05 Boston Biomedical, Inc. Composition d'arn duplex asymétrique utile comme mimétique ou inhibiteur de micro arn
WO2009033033A2 (fr) 2007-09-06 2009-03-12 Boston Biomedical, Inc. Compositions d'inhibiteurs de kinase et leur utilisation pour le traitement du cancer et d'autres maladies liées aux kinases
WO2009036099A1 (fr) 2007-09-10 2009-03-19 Boston Biomedical, Inc. Groupe innovant d'inhibiteurs de la voie stat3 et d'inhibiteurs de la voie des cellules souches cancéreuses
WO2009100140A1 (fr) 2008-02-04 2009-08-13 Medarex, Inc. Anticorps anti-clta-4 avec blocage réduit de la liaison de ctla-4 à b7 et leurs utilisations
US20090239936A1 (en) 2006-05-15 2009-09-24 Yoshikazu Sugimoto Prophylactic and Therapeutic Agent for Cancer
WO2009143372A2 (fr) 2008-05-21 2009-11-26 Intradigm Corporation Compositions comportant des arnsi des gènes a-raf, b-raf, et c-raf et leurs procédés d’utilisation
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
WO2010042433A1 (fr) 2008-10-06 2010-04-15 Bristol-Myers Squibb Company Combinaison d'anticorps cd137 et d'anticorps ctla-4 pour le traitement de maladies prolifératives
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
WO2011011027A1 (fr) 2009-07-20 2011-01-27 Bristol-Myers Squibb Company Combinaison d’un anticorps anti-ctla-4 avec divers régimes de traitement pour le traitement synergique de maladies prolifératives
WO2011045704A1 (fr) 2009-10-12 2011-04-21 Pfizer Inc. Traitement d'un cancer
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
WO2011116398A1 (fr) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Nouveaux composés et compositions pour le ciblage de cellules souches cancéreuses
WO2011116399A1 (fr) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Nouveaux procédés de ciblage des cellules souches du cancer
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2012018881A2 (fr) 2010-08-03 2012-02-09 Alnylam Pharmaceuticals, Inc. Procédés et compositions pour la régulation d'arn
US8119129B2 (en) 2008-08-01 2012-02-21 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with dasatinib for the treatment of proliferative diseases
WO2012027536A1 (fr) 2010-08-26 2012-03-01 Bristol-Myers Squibb Company Combinaison d'anticorps anti-ctla4 avec des inhibiteurs de braf pour le traitement synergique de maladies prolifératives
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2013019620A2 (fr) 2011-07-29 2013-02-07 Glaxosmithkline Llc Méthode de traitement du cancer au moyen d'une combinaison contenant un inhibiteur de braf, un inhibiteur de mek et un anticorps anti-ctla-4
WO2013059320A1 (fr) 2011-10-17 2013-04-25 Board Of Regents, The University Of Texas System Traitement de cancers caractérisés par la présence d'une protéine npm1 mutante au moyen d'inhibiteurs de mek
US8449886B2 (en) 2008-01-08 2013-05-28 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with tubulin modulating agents for the treatment of proliferative diseases
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
WO2013142796A2 (fr) 2012-03-23 2013-09-26 Bristol-Myers Squibb Company Procédés de traitements au moyen d'anticorps anti-ctla4
US8609831B2 (en) 2002-09-16 2013-12-17 University Of Southern California RNA-mediated gene modulation
US8686119B2 (en) 2011-07-24 2014-04-01 Curetech Ltd. Variants of humanized immunomodulatory monoclonal antibodies
WO2014047973A1 (fr) 2012-09-28 2014-04-03 武汉纽斯特生物技术有限公司 Anticorps monoclonaux reconnaissant, de façon spécifique, les protéines mutantes b-raf, leur procédé de préparation et leur utilisation
WO2014066532A1 (fr) 2012-10-23 2014-05-01 Bristol-Myers Squibb Company Association d'anticorps anti-kir et anti-ctla-4 pour le traitement du cancer
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
US8747847B2 (en) 2008-02-11 2014-06-10 Curetech Ltd. Monoclonal antibodies for tumor treatment
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
US8784815B2 (en) 1999-08-24 2014-07-22 Medarex, L.L.C. Human CTLA-4 antibodies and their uses
WO2014123935A1 (fr) 2013-02-05 2014-08-14 1Globe Health Institute Llc Nanoparticules biodégradables et cliniquement compatibles en tant que véhicules d'administration de médicament
WO2014152588A1 (fr) 2013-03-15 2014-09-25 Araxes Pharma Llc Inhibiteurs covalents de k-ras g12c
WO2014160401A1 (fr) 2013-03-13 2014-10-02 Boston Biomedical, Inc. Dérivés de 3-(aryl ou hétéroaryl)méthylèneindolin-2-one en tant qu'inhibiteurs de kinases de la voie des cellules souches cancéreuses pour le traitement de cancer
WO2014169078A2 (fr) 2013-04-09 2014-10-16 Boston Biomedical, Inc. Procédés de traitement du cancer
WO2014190356A2 (fr) 2013-05-24 2014-11-27 Amplimmune, Inc. Anticorps anti-b7-h5 et leurs utilisations
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2014197849A2 (fr) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Anticorps anti-c10orf54 et leurs utilisations
WO2015001045A2 (fr) 2013-07-03 2015-01-08 Universite Pierre Et Marie Curie (Paris 6) Peptides ras et raf pro-apoptotiques
US8977803B2 (en) 2011-11-21 2015-03-10 Western Digital Technologies, Inc. Disk drive data caching using a multi-tiered memory
WO2015034820A1 (fr) 2013-09-04 2015-03-12 Bristol-Myers Squibb Company Composés utiles comme immunomodulateurs
US20150087581A1 (en) 2010-06-25 2015-03-26 Aurigene Discovery Technologies Limited Immunosuppression Modulating Compounds
WO2015050844A1 (fr) 2013-10-01 2015-04-09 Dana-Farber Cancer Institute, Inc. Procédés de traitement anticancéreux par des composés apparentés à l'atovaquone
US20150125491A1 (en) 2012-03-29 2015-05-07 Aurigene Discovery Technologies Limited Immunomodulating Cyclic Compounds
US9044442B2 (en) 2012-03-07 2015-06-02 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
WO2015097536A2 (fr) 2013-12-24 2015-07-02 Janssen Pharmaceutical Nv Anticorps et fragments anti-vista
US20150197571A1 (en) 2012-08-03 2015-07-16 Dana-Farber Cancer Institute, Inc. Single Agent Anti-PD-L1 and PD-L2 Dual Binding Antibodies and Methods of Use
US20150203579A1 (en) 2014-01-23 2015-07-23 Regeneron Pharmaceuticals, Inc. Human Antibodies to PD-1
US9096642B2 (en) 2011-06-08 2015-08-04 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
US9115122B2 (en) 2012-12-20 2015-08-25 University Of Maryland, Baltimore Non-ATP dependent inhibitors of extracellular signal-regulated kinase (ERK)
WO2015134605A1 (fr) 2014-03-05 2015-09-11 Bristol-Myers Squibb Company Traitement du cancer du rein à l'aide d'une combinaison d'un anticorps anti-pd-1 et d'un autre agent anticancéreux
WO2015139044A1 (fr) 2014-03-14 2015-09-17 Boston Biomedical, Inc. Compositions d'arn interférent asymétrique qui réduisent au silence k-ras et procédés d'utilisation de celles-ci
US9150530B2 (en) 2011-03-04 2015-10-06 Zhoushan Haizhongzhou Xinsheng Pharmaceuticals Co., Ltd. Esters of 4, 9-dihydroxy-naphtho [2, 3-b] furans for disease therapies
US9187454B2 (en) 2013-03-13 2015-11-17 Boston Biomedical, Inc. Inhibitors of kinases and cancer stem cells, and methods of preparation and use thereof
WO2015173267A1 (fr) 2014-05-13 2015-11-19 Medimmune Limited Anticorps anti-b7-h1 et anti-ctla -4 pour le traitement du cancer du poumon non à petites cellules
US9205148B2 (en) 2011-04-20 2015-12-08 Medimmune, Llc Antibodies and other molecules that bind B7-H1 and PD-1
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
US9220776B2 (en) 2011-03-31 2015-12-29 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor PD-1 and related treatments
US9233940B2 (en) 2013-09-06 2016-01-12 Aurigene Discovery Technologies Limited Cyclic peptidomimetic compounds as immunomodulators
US9255147B2 (en) 2004-11-05 2016-02-09 Mayo Foundation For Medical Education & Research Recombinantly produced antibody
US9260470B2 (en) 2009-11-04 2016-02-16 Sungkyunkwan University Foundation For Corporate Collaboration SiRNA structure for minimizing off-target effects caused by antisense strands, and use thereof
WO2016030455A1 (fr) 2014-08-28 2016-03-03 Medimmune Limited Anticorps anti-b7-h1 et anti-ctla -4 pour le traitement du cancer du poumon non à petites cellules
US20160075783A1 (en) 2013-05-02 2016-03-17 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2016049524A1 (fr) 2014-09-25 2016-03-31 Araxes Pharma Llc Inhibiteurs de protéines mutantes kras g12c
US20160113901A1 (en) 2013-09-06 2016-04-28 Aurigene Discovery Technologies Limited Cyclic Peptidomimetic Compounds as Immunomodulators
WO2016094837A2 (fr) 2014-12-11 2016-06-16 Igenica Biotherapeutics, Inc. Anticorps anti-c10orf54 et leurs utilisations
US20160194295A1 (en) 2013-09-06 2016-07-07 Aurigene Discovery Technologies Limited 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators
WO2016123378A1 (fr) 2015-01-30 2016-08-04 Kim Hyungsoo Thérapie anticancéreuse utilisant un inhibiteur de jak en combinaison avec des inhibiteurs de mapk
US20160222120A1 (en) 2013-09-11 2016-08-04 Medlmmune Limited Anti-B7-H1 Antibodies for Treating Tumors
WO2016137985A1 (fr) 2015-02-26 2016-09-01 Merck Patent Gmbh Inhibiteurs de pd-1/pd-l1 pour le traitement du cancer
WO2016142835A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés cycliques thérapeutiques utilisés en tant qu'immunomodulateurs
WO2016142833A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 1,2,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
WO2016142852A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 1,3,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
WO2016142894A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés de 1,3,4-oxadiazole et thiadiazole substitués en position 3 utilisés en tant qu'immunomodulateurs
WO2016142886A2 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 3-substitué -1,2,4-oxadiazole et thiadiazole utilisés comme immunomodulateurs
US9447170B2 (en) 2008-04-18 2016-09-20 Avacta Life Sciences Limited Modified Stefin A scaffold proteins

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7514416B2 (en) * 2004-04-27 2009-04-07 Japan Health Sciences Foundation Interferon alpha and antisense K-ras RNA combination gene therapy
AR091004A1 (es) * 2012-05-09 2014-12-30 Gradalis Inc Arn en horquilla corta bifuncional (bi-arnhc) especifico de mutaciones de nucleotidos individuales en k-ras
WO2014195852A1 (fr) * 2013-06-03 2014-12-11 Glaxosmithkline Intellectual Property (No.2) Limited Combinaisons d'un anticorps anti-pd-l1 et d'un inhibiteur de mek et/ou d'un inhibiteur de braf
WO2015164818A1 (fr) * 2014-04-25 2015-10-29 Strike Bio, Inc. Arni à cibles multiples permettant le traitement de cancers
PE20171067A1 (es) * 2014-10-14 2017-07-24 Novartis Ag Moleculas de anticuerpo que se unen a pd-l1 y usos de las mismas

Patent Citations (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4657760A (en) 1979-03-20 1987-04-14 Ortho Pharmaceutical Corporation Methods and compositions using monoclonal antibody to human T cells
US4898932A (en) 1985-01-29 1990-02-06 E. I. Du Pont De Nemours And Company Monoclonal antibodies reactive with activated and oncogenic ras p21 proteins
US5084380A (en) 1985-01-29 1992-01-28 Applied Biotechnology Monoclonal antibodies reactive with activated and oncogenic ras p21 proteins
US5206344A (en) 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
US4820631A (en) 1986-07-30 1989-04-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Deletion mutants and monoclonal antibodies against ras proteins
US5081230A (en) 1987-07-08 1992-01-14 E. I. Dupont Denemours And Company Monoclonal antibodies reactive with normal and oncogenic forms of the ras p21 protein
US5028527A (en) 1988-02-22 1991-07-02 Applied Bio Technology Monoclonal antibodies against activated ras proteins with amino acid mutations at position 13 of the protein
US5112737A (en) 1988-02-22 1992-05-12 Applied Biotechnology, Inc. Monoclonal antibodies against activated ras proteins with amino acid mutations at position 13 of the protein
WO1990005719A1 (fr) 1988-11-23 1990-05-31 British Bio-Technology Limited Inhibiteurs de collagenase a base d'acide hydroxamique
US5225347A (en) 1989-09-25 1993-07-06 Innovir Laboratories, Inc. Therapeutic ribozyme compositions and expression vectors
US5225212A (en) 1989-10-20 1993-07-06 Liposome Technology, Inc. Microreservoir liposome composition and method
US5965539A (en) 1993-05-18 1999-10-12 Univeristy Of Pittsburgh Inhibitors of prenyl transferases
WO1996027583A1 (fr) 1995-03-08 1996-09-12 Pfizer Inc. Derives de l'acide arylsulfonylamino hydroxamique
US5863949A (en) 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
WO1996033172A1 (fr) 1995-04-20 1996-10-24 Pfizer Inc. Derives d'acide hydroxamique arylsufonyle en tant qu'inhibiteurs de mmp et de tnf
US5861510A (en) 1995-04-20 1999-01-19 Pfizer Inc Arylsulfonyl hydroxamic acid derivatives as MMP and TNF inhibitors
US5693773A (en) 1995-06-07 1997-12-02 Hybridon Incorporated Triplex-forming antisense oligonucleotides having abasic linkers targeting nucleic acids comprising mixed sequences of purines and pyrimidines
US6107094A (en) 1996-06-06 2000-08-22 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
WO1998003516A1 (fr) 1996-07-18 1998-01-29 Pfizer Inc. Composes a base de phosphinate inhibiteurs des metalloproteases matricielles
WO1998007697A1 (fr) 1996-08-23 1998-02-26 Pfizer Inc. Derives de l'acide arylsulfonylamino hydroxamique
WO1998030566A1 (fr) 1997-01-06 1998-07-16 Pfizer Inc. Derives de sulfone cyclique
WO1998033768A1 (fr) 1997-02-03 1998-08-06 Pfizer Products Inc. Derives d'acide arylsulfonylaminohydroxamique
WO1998034915A1 (fr) 1997-02-07 1998-08-13 Pfizer Inc. Derives du n-hxdroxy-beta-sulfonyl-propionamide et leur utilisation comme inhibiteurs des metalloproteases matrices
WO1998034918A1 (fr) 1997-02-11 1998-08-13 Pfizer Inc. Derives de l'acide arylsulfonylhydroxamique
WO1999029667A1 (fr) 1997-12-05 1999-06-17 Pfizer Limited Derives d'acide hydroxamique utilises comme inhibiteurs de metalloproteases matricielles
WO1999052889A1 (fr) 1998-04-10 1999-10-21 Pfizer Products Inc. Hydroxamides de l'acide (4-arylsulfonylamino)-tetrahydropyrane-4-carboxylique
WO1999052910A1 (fr) 1998-04-10 1999-10-21 Pfizer Products Inc. Derives bicycliques de l'acide hydroxamique
US7824679B2 (en) 1998-12-23 2010-11-02 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US7132281B2 (en) 1998-12-23 2006-11-07 Amgen Fremont Inc. Methods and host cells for producing human monoclonal antibodies to CTLA-4
US8883984B2 (en) 1998-12-23 2014-11-11 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US8491895B2 (en) 1998-12-23 2013-07-23 Amgen Fremont Inc. Methods of treating cancer with human monoclonal antibodies to CTLA-4
US8143379B2 (en) 1998-12-23 2012-03-27 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US7411057B2 (en) 1998-12-23 2008-08-12 Amgen Fremont Inc. Nucleic acids encoding human monoclonal antibodies to CTLA-4
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2001014424A2 (fr) 1999-08-24 2001-03-01 Medarex, Inc. Anticorps contre l'antigene ctla-4 humain et utilisation
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
US8784815B2 (en) 1999-08-24 2014-07-22 Medarex, L.L.C. Human CTLA-4 antibodies and their uses
US6124133A (en) 1999-10-15 2000-09-26 Isis Pharmaceuticals Inc. Antisense inhibition of fra-1 expression
US7056704B2 (en) 2000-12-01 2006-06-06 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
WO2003086459A1 (fr) 2002-04-12 2003-10-23 Medarex, Inc. Methodes de traitement dans lesquelles sont utilises des anticorps du ctla-4
US7332582B2 (en) 2002-05-23 2008-02-19 Curetech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
US8609831B2 (en) 2002-09-16 2013-12-17 University Of Southern California RNA-mediated gene modulation
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
WO2005092380A2 (fr) 2004-03-26 2005-10-06 Pfizer Products Inc Utilisations d'anticorps anti-ctla-4
WO2006048749A1 (fr) 2004-11-04 2006-05-11 Pfizer Products Inc. Anticorps ctla-4 et inhibiteur de l'aromatase ou traitement combine du cancer du sein
US20160122431A1 (en) 2004-11-05 2016-05-05 Larry R. Pease B7-dc binding antibody
US9255147B2 (en) 2004-11-05 2016-02-09 Mayo Foundation For Medical Education & Research Recombinantly produced antibody
WO2006096491A2 (fr) 2005-03-08 2006-09-14 Pharmacia & Upjohn Company Llc Compositions d'anticorps anti-ctla-4
WO2006101691A1 (fr) 2005-03-23 2006-09-28 Pfizer Products Inc. Traitement du cancer de la prostate au moyen d'anticorps anti-ctla4 et d'une hormonotherapie
WO2006101692A1 (fr) 2005-03-23 2006-09-28 Pfizer Products Inc. Association d'anticorps anti-ctla4 et d'indolinone dans le traitement du cancer
US8779105B2 (en) 2005-05-09 2014-07-15 Medarex, L.L.C. Monoclonal antibodies to programmed death 1 (PD-1)
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US9358289B2 (en) 2005-05-09 2016-06-07 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies in combination with anti-CTLA-4 antibodies
US9387247B2 (en) 2005-05-09 2016-07-12 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to programmed death 1 (PD-1)
US9273135B2 (en) 2005-07-01 2016-03-01 E. R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US9102725B2 (en) 2005-07-01 2015-08-11 E. R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007056539A2 (fr) 2005-11-08 2007-05-18 Medarex, Inc. Prevention et traitement de l'enterocolite associee a un traitement par anticorps anti-ctla-4
WO2007067959A2 (fr) 2005-12-07 2007-06-14 Medarex, Inc. Regimes d'escalade de dose d'anticorps ctla-4
US9062111B2 (en) 2005-12-07 2015-06-23 Medarex, L.L.C. CTLA-4 antibody dosage escalation regimens
US8110194B2 (en) 2005-12-07 2012-02-07 Medarex, Inc. CTLA-4 antibody dosage escalation regimens
WO2007113648A2 (fr) 2006-04-05 2007-10-11 Pfizer Products Inc. Polythérapie à base d'un anticorps anti-ctla4
US20090239936A1 (en) 2006-05-15 2009-09-24 Yoshikazu Sugimoto Prophylactic and Therapeutic Agent for Cancer
WO2008109142A2 (fr) 2007-03-07 2008-09-12 Cold Spring Harbor Laboratory ARNpi ET UTILISATIONS CORRESPONDANTES
US8900587B2 (en) 2007-06-18 2014-12-02 Merck Sharp & Dohme Corp. Antibodies to human programmed death receptor PD-1
US8952136B2 (en) 2007-06-18 2015-02-10 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor PD-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2009029688A2 (fr) 2007-08-27 2009-03-05 Boston Biomedical, Inc. Compositions d'arn interférent asymétrique et leurs utilisations
WO2009029690A1 (fr) 2007-08-27 2009-03-05 Boston Biomedical, Inc. Composition d'arn duplex asymétrique utile comme mimétique ou inhibiteur de micro arn
US9328345B2 (en) 2007-08-27 2016-05-03 1 Globe Health Institute Llc Compositions of asymmetric interfering RNA and uses thereof
WO2009033033A2 (fr) 2007-09-06 2009-03-12 Boston Biomedical, Inc. Compositions d'inhibiteurs de kinase et leur utilisation pour le traitement du cancer et d'autres maladies liées aux kinases
US8299106B2 (en) 2007-09-06 2012-10-30 Boston Biomedical, Inc. Compositions of kinase inhibitors and their use for treatment of cancer and other diseases related to kinases
WO2009036101A1 (fr) 2007-09-10 2009-03-19 Boston Biomedical, Inc. Compositions et procédés nouveaux pour le traitement du cancer
WO2009036099A1 (fr) 2007-09-10 2009-03-19 Boston Biomedical, Inc. Groupe innovant d'inhibiteurs de la voie stat3 et d'inhibiteurs de la voie des cellules souches cancéreuses
US20120252763A1 (en) 2007-09-10 2012-10-04 Boston Biomedical, Inc. Novel group of stat3 pathway inhibitors and cancer stem cell pathway inhibitors
US8449886B2 (en) 2008-01-08 2013-05-28 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with tubulin modulating agents for the treatment of proliferative diseases
WO2009100140A1 (fr) 2008-02-04 2009-08-13 Medarex, Inc. Anticorps anti-clta-4 avec blocage réduit de la liaison de ctla-4 à b7 et leurs utilisations
US8747847B2 (en) 2008-02-11 2014-06-10 Curetech Ltd. Monoclonal antibodies for tumor treatment
US9309308B2 (en) 2008-02-11 2016-04-12 Curetech Ltd. Monoclonal antibodies for tumor treatment
US9447170B2 (en) 2008-04-18 2016-09-20 Avacta Life Sciences Limited Modified Stefin A scaffold proteins
WO2009143372A2 (fr) 2008-05-21 2009-11-26 Intradigm Corporation Compositions comportant des arnsi des gènes a-raf, b-raf, et c-raf et leurs procédés d’utilisation
US8685394B2 (en) 2008-08-01 2014-04-01 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with diverse therapeutic regimens for the synergistic treatment of proliferative diseases
US9320811B2 (en) 2008-08-01 2016-04-26 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with diverse therapeutic regimens for the synergistic treatment of proliferative diseases
US8119129B2 (en) 2008-08-01 2012-02-21 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with dasatinib for the treatment of proliferative diseases
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
US20130291136A1 (en) 2008-09-26 2013-10-31 Emory University Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US20160137731A1 (en) 2008-09-26 2016-05-19 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2010042433A1 (fr) 2008-10-06 2010-04-15 Bristol-Myers Squibb Company Combinaison d'anticorps cd137 et d'anticorps ctla-4 pour le traitement de maladies prolifératives
US8475790B2 (en) 2008-10-06 2013-07-02 Bristol-Myers Squibb Company Combination of CD137 antibody and CTLA-4 antibody for the treatment of proliferative diseases
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2011011027A1 (fr) 2009-07-20 2011-01-27 Bristol-Myers Squibb Company Combinaison d’un anticorps anti-ctla-4 avec divers régimes de traitement pour le traitement synergique de maladies prolifératives
WO2011045704A1 (fr) 2009-10-12 2011-04-21 Pfizer Inc. Traitement d'un cancer
US9260470B2 (en) 2009-11-04 2016-02-16 Sungkyunkwan University Foundation For Corporate Collaboration SiRNA structure for minimizing off-target effects caused by antisense strands, and use thereof
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
WO2011116399A1 (fr) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Nouveaux procédés de ciblage des cellules souches du cancer
WO2011116398A1 (fr) 2010-03-19 2011-09-22 Boston Biomedical, Inc. Nouveaux composés et compositions pour le ciblage de cellules souches cancéreuses
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
US20150087581A1 (en) 2010-06-25 2015-03-26 Aurigene Discovery Technologies Limited Immunosuppression Modulating Compounds
WO2012018881A2 (fr) 2010-08-03 2012-02-09 Alnylam Pharmaceuticals, Inc. Procédés et compositions pour la régulation d'arn
WO2012027536A1 (fr) 2010-08-26 2012-03-01 Bristol-Myers Squibb Company Combinaison d'anticorps anti-ctla4 avec des inhibiteurs de braf pour le traitement synergique de maladies prolifératives
US9150530B2 (en) 2011-03-04 2015-10-06 Zhoushan Haizhongzhou Xinsheng Pharmaceuticals Co., Ltd. Esters of 4, 9-dihydroxy-naphtho [2, 3-b] furans for disease therapies
US9220776B2 (en) 2011-03-31 2015-12-29 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor PD-1 and related treatments
US9205148B2 (en) 2011-04-20 2015-12-08 Medimmune, Llc Antibodies and other molecules that bind B7-H1 and PD-1
US20160130348A1 (en) 2011-04-20 2016-05-12 Medimmune, Llc Antibodies and other molecules that bind b7-h1 and pd-1
US9096642B2 (en) 2011-06-08 2015-08-04 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
US9416175B2 (en) 2011-07-24 2016-08-16 Curetech Ltd. Variants of humanized immunomodulatory monoclonal antibodies
US8686119B2 (en) 2011-07-24 2014-04-01 Curetech Ltd. Variants of humanized immunomodulatory monoclonal antibodies
WO2013019620A2 (fr) 2011-07-29 2013-02-07 Glaxosmithkline Llc Méthode de traitement du cancer au moyen d'une combinaison contenant un inhibiteur de braf, un inhibiteur de mek et un anticorps anti-ctla-4
WO2013059320A1 (fr) 2011-10-17 2013-04-25 Board Of Regents, The University Of Texas System Traitement de cancers caractérisés par la présence d'une protéine npm1 mutante au moyen d'inhibiteurs de mek
US8977803B2 (en) 2011-11-21 2015-03-10 Western Digital Technologies, Inc. Disk drive data caching using a multi-tiered memory
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US9044442B2 (en) 2012-03-07 2015-06-02 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
WO2013142796A2 (fr) 2012-03-23 2013-09-26 Bristol-Myers Squibb Company Procédés de traitements au moyen d'anticorps anti-ctla4
US20150125491A1 (en) 2012-03-29 2015-05-07 Aurigene Discovery Technologies Limited Immunomodulating Cyclic Compounds
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
US20150197571A1 (en) 2012-08-03 2015-07-16 Dana-Farber Cancer Institute, Inc. Single Agent Anti-PD-L1 and PD-L2 Dual Binding Antibodies and Methods of Use
WO2014047973A1 (fr) 2012-09-28 2014-04-03 武汉纽斯特生物技术有限公司 Anticorps monoclonaux reconnaissant, de façon spécifique, les protéines mutantes b-raf, leur procédé de préparation et leur utilisation
WO2014066532A1 (fr) 2012-10-23 2014-05-01 Bristol-Myers Squibb Company Association d'anticorps anti-kir et anti-ctla-4 pour le traitement du cancer
US9115122B2 (en) 2012-12-20 2015-08-25 University Of Maryland, Baltimore Non-ATP dependent inhibitors of extracellular signal-regulated kinase (ERK)
WO2014123935A1 (fr) 2013-02-05 2014-08-14 1Globe Health Institute Llc Nanoparticules biodégradables et cliniquement compatibles en tant que véhicules d'administration de médicament
US9187454B2 (en) 2013-03-13 2015-11-17 Boston Biomedical, Inc. Inhibitors of kinases and cancer stem cells, and methods of preparation and use thereof
WO2014160401A1 (fr) 2013-03-13 2014-10-02 Boston Biomedical, Inc. Dérivés de 3-(aryl ou hétéroaryl)méthylèneindolin-2-one en tant qu'inhibiteurs de kinases de la voie des cellules souches cancéreuses pour le traitement de cancer
WO2014152588A1 (fr) 2013-03-15 2014-09-25 Araxes Pharma Llc Inhibiteurs covalents de k-ras g12c
WO2014169078A2 (fr) 2013-04-09 2014-10-16 Boston Biomedical, Inc. Procédés de traitement du cancer
US20160075783A1 (en) 2013-05-02 2016-03-17 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
US20160096891A1 (en) 2013-05-24 2016-04-07 Medimmune, Llc Anti-b7-h5 antibodies and their uses
WO2014190356A2 (fr) 2013-05-24 2014-11-27 Amplimmune, Inc. Anticorps anti-b7-h5 et leurs utilisations
WO2014197849A2 (fr) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Anticorps anti-c10orf54 et leurs utilisations
WO2015001045A2 (fr) 2013-07-03 2015-01-08 Universite Pierre Et Marie Curie (Paris 6) Peptides ras et raf pro-apoptotiques
WO2015034820A1 (fr) 2013-09-04 2015-03-12 Bristol-Myers Squibb Company Composés utiles comme immunomodulateurs
US9233940B2 (en) 2013-09-06 2016-01-12 Aurigene Discovery Technologies Limited Cyclic peptidomimetic compounds as immunomodulators
US20160194295A1 (en) 2013-09-06 2016-07-07 Aurigene Discovery Technologies Limited 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators
US20160113901A1 (en) 2013-09-06 2016-04-28 Aurigene Discovery Technologies Limited Cyclic Peptidomimetic Compounds as Immunomodulators
US20160222120A1 (en) 2013-09-11 2016-08-04 Medlmmune Limited Anti-B7-H1 Antibodies for Treating Tumors
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
US9217034B2 (en) 2013-09-13 2015-12-22 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2015050844A1 (fr) 2013-10-01 2015-04-09 Dana-Farber Cancer Institute, Inc. Procédés de traitement anticancéreux par des composés apparentés à l'atovaquone
WO2015097536A2 (fr) 2013-12-24 2015-07-02 Janssen Pharmaceutical Nv Anticorps et fragments anti-vista
US20150203579A1 (en) 2014-01-23 2015-07-23 Regeneron Pharmaceuticals, Inc. Human Antibodies to PD-1
WO2015134605A1 (fr) 2014-03-05 2015-09-11 Bristol-Myers Squibb Company Traitement du cancer du rein à l'aide d'une combinaison d'un anticorps anti-pd-1 et d'un autre agent anticancéreux
WO2015139044A1 (fr) 2014-03-14 2015-09-17 Boston Biomedical, Inc. Compositions d'arn interférent asymétrique qui réduisent au silence k-ras et procédés d'utilisation de celles-ci
WO2015173267A1 (fr) 2014-05-13 2015-11-19 Medimmune Limited Anticorps anti-b7-h1 et anti-ctla -4 pour le traitement du cancer du poumon non à petites cellules
WO2016030455A1 (fr) 2014-08-28 2016-03-03 Medimmune Limited Anticorps anti-b7-h1 et anti-ctla -4 pour le traitement du cancer du poumon non à petites cellules
WO2016049524A1 (fr) 2014-09-25 2016-03-31 Araxes Pharma Llc Inhibiteurs de protéines mutantes kras g12c
WO2016094837A2 (fr) 2014-12-11 2016-06-16 Igenica Biotherapeutics, Inc. Anticorps anti-c10orf54 et leurs utilisations
WO2016123378A1 (fr) 2015-01-30 2016-08-04 Kim Hyungsoo Thérapie anticancéreuse utilisant un inhibiteur de jak en combinaison avec des inhibiteurs de mapk
WO2016137985A1 (fr) 2015-02-26 2016-09-01 Merck Patent Gmbh Inhibiteurs de pd-1/pd-l1 pour le traitement du cancer
WO2016142835A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés cycliques thérapeutiques utilisés en tant qu'immunomodulateurs
WO2016142833A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 1,2,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
WO2016142852A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 1,3,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
WO2016142894A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés de 1,3,4-oxadiazole et thiadiazole substitués en position 3 utilisés en tant qu'immunomodulateurs
WO2016142886A2 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 3-substitué -1,2,4-oxadiazole et thiadiazole utilisés comme immunomodulateurs

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"Antibodies: A Laboratory Manual", 2014, COLD SPRING HARBOR LABORATORY PRESS
"Remington: The Science and Practice of Pharmacy, Twentieth Edition", LIPPINCOTT WILLIAMS & WILKINS
AUSUBEL ET AL.: "Current Protocols", JOHN WILEY & SONS, INC., pages: 2000 - 2016
BRAHMER ET AL., N. ENGL. J. MED., vol. 366, 2012, pages 2455 - 65
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1374853-91-4
DAR ET AL.: "siRNAmod: A database of experimentally validated chemically modified siRNAs", SCI. REP., vol. 6, 2016, pages 20031
J CLIN ONCOL, vol. 34, 2016
LIPSON ET AL., SCMIN. ONCOL., vol. 42, no. 4, 2015, pages 587 - 600
MR. GREEN; J. SAMBROOK: "Molecular Cloning: A Laboratory Manual", 2012, COLD SPRING HARBOR LABORATORY PRESS
RIO ET AL.: "A Laboratory Manual", 2011, COLD SPRING HARBOR LABORATORY PRESS
STEPHEN, CANCER CELL, vol. 25, no. 3, 2014, pages 272 - 281
TANIGAWA ET AL., CANCER RES, vol. 42, 1982, pages 2159 - 2164
WEISCNTHAL ET AL., CANCER TREAT REP, vol. 69, 1985, pages 615 - 632
WEISENTHAL ET AL., CANCER RES 1 84, vol. 94, pages 161 - 173
WEISENTHAL ET AL.: "Drug Resistance in Leukemia and Lymphoma", vol. 1993, 1994, HARWCOD ACADEMIC PUBLISHERS, pages: 41 - 432
WU ET AL., DRUG DISCOVERY TODAY, vol. 21, no. 1, 2016, pages 5 - 10
ZOU ET AL., SCI. TRANSL. MED., vol. 8, no. 328, 2016, pages 328rv4

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11840533B2 (en) 2017-12-01 2023-12-12 The Board Of Trustees Of The University Of Illinois Pyridinone-based epigenetic modifiers and uses thereof
WO2020014650A1 (fr) * 2018-07-13 2020-01-16 Memorial Sloan Kettering Cancer Center Méthodes d'utilisation d'inhibiteurs pharmacologiques de la signalisation des cytokines de type 2 pour traiter ou prévenir le cancer du pancréas
WO2020081905A1 (fr) * 2018-10-18 2020-04-23 Jounce Therapeutics, Inc Méthodes de traitement du cancer
US20210353750A1 (en) * 2018-10-18 2021-11-18 Jounce Therapeutics, Inc. Methods of Treating Cancer
WO2020205473A1 (fr) * 2019-03-29 2020-10-08 Decerna Pharmaceuticals, Inc. Compositions et procédés pour le traitement de maladies et troubles associés au kras
CN113924365A (zh) * 2019-03-29 2022-01-11 迪克纳制药公司 用于治疗kras相关疾病或病症的组合物和方法
WO2021016228A1 (fr) * 2019-07-22 2021-01-28 H. Lee Moffitt Cancer Center And Research Institute Inc. Polythérapie pour le traitement de cancers à mutant ras
WO2021061749A1 (fr) * 2019-09-24 2021-04-01 Mirati Therapeutics, Inc. Polythérapies
CN114761012A (zh) * 2019-09-24 2022-07-15 米拉蒂治疗股份有限公司 组合疗法
US11890285B2 (en) 2019-09-24 2024-02-06 Mirati Therapeutics, Inc. Combination therapies
WO2023087012A1 (fr) * 2021-11-15 2023-05-19 The Board Of Trustees Of The University Of Illinois Méthode de traitement d'un cancer lié à une mutation ras
WO2023172940A1 (fr) * 2022-03-08 2023-09-14 Revolution Medicines, Inc. Méthodes de traitement du cancer du poumon réfractaire immunitaire

Also Published As

Publication number Publication date
WO2018098352A3 (fr) 2018-07-05

Similar Documents

Publication Publication Date Title
WO2018098352A2 (fr) Ciblage d'expression du point de contrôle immunitaire induit par kras
Hou et al. Navigating CAR-T cells through the solid-tumour microenvironment
US20210145977A1 (en) Methods and pharmaceutical composition for the treatment of cancer
ES2638521T5 (es) Composiciones y métodos para anticuerpos y proteínas de fusión inmunomoduladores dirigidos contra dianas
JP2018532810A (ja) がんの処置のための腫瘍抑制因子遺伝子治療および免疫チェックポイント治療を含む組成物
JP2020510624A (ja) がんおよび感染性疾患の治療および予防のための、ウイルス遺伝子治療および免疫チェックポイント阻害剤を含む方法および組成物
US20200157225A1 (en) Inhibitor of suv39h1 histone methyltransferase for use in cancer combination therapy
TW201717935A (zh) 用於治療癌症的組成物和方法
KR20180102628A (ko) 면역조절제의 조합물에 의한 암의 치료
JP7148406B2 (ja) 免疫チェックポイントモジュレーターとのt細胞リダイレクティング多機能抗体の組み合わせとその使用
US20210169880A1 (en) Control and modulation of the function of gene-modified chimeric antigen receptor t cells with dasatinib and other tyrosine kinase inhibitors
KR20180015269A (ko) 암 치료에 사용하기 위한 체크포인트 저해제와 마이코박테리움 전체 세포
KR20210106531A (ko) 암 치료를 위한 조성물 및 방법
US20200405853A1 (en) Inhibitor of setdb1 histone methyltransferase for use in cancer combination therapy
JP7407452B2 (ja) がんの治療及び/又は予防のための医薬
EP4353741A1 (fr) Cellules tueuses naturelles à double inactivation
WO2023212566A1 (fr) Compositions et procédés pour empêcher l'épuisement des lymphocytes t
US20200163988A1 (en) Immunosuppression-Reverting Oligonucleotides Inhibiting the Expression of IDO
US20190298666A1 (en) Promotion of t lymphocyte proliferation
EP4017876A1 (fr) Matériaux et procédés pour activer des réponses de lymphocytes t spécifiques d'un antigène
CN116390936A (zh) 嵌合tim受体及其用途
NZ750663A (en) Compositions and methods for cancer immunotherapy
NZ750663B2 (en) Compositions and methods for cancer immunotherapy

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: 17817465

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17817465

Country of ref document: EP

Kind code of ref document: A2