WO2023230554A1 - Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer - Google Patents

Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer Download PDF

Info

Publication number
WO2023230554A1
WO2023230554A1 PCT/US2023/067469 US2023067469W WO2023230554A1 WO 2023230554 A1 WO2023230554 A1 WO 2023230554A1 US 2023067469 W US2023067469 W US 2023067469W WO 2023230554 A1 WO2023230554 A1 WO 2023230554A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
subject
amount
braf
msi
Prior art date
Application number
PCT/US2023/067469
Other languages
French (fr)
Inventor
Adam Brent SCHAYOWITZ
Xiaosong Zhang
Ashwin Mohan GOLLERKERI
Original Assignee
Pfizer Inc.
Msd International Gmbh
Eli Lilly And Company
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 Pfizer Inc., Msd International Gmbh, Eli Lilly And Company filed Critical Pfizer Inc.
Publication of WO2023230554A1 publication Critical patent/WO2023230554A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • C07K16/2818Immunoglobulins [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 CD28 or CD152
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule

Definitions

  • This application incorporates the contents of a 23.6 kb xml file created on May 25, 2023 and named “SequenceListing.xml,” which is the sequence listing for this application.
  • the present invention relates to combination therapies useful for the treatment of previously untreated BRAF V600E-mutant, microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) colorectal cancer.
  • the invention relates to a combination therapy comprising a BRAF inhibitor, an EGFR inhibitor, and an antagonist of a Programmed Death 1 protein (PD-1 ).
  • PD-1 Programmed Death 1 protein
  • the invention also relates to associated methods of treatment, pharmaceutical compositions, and pharmaceutical uses.
  • MSI-H/dMMR microsatellite high/ mismatch repair deficient
  • MSI microsatellite instability
  • MSI-H microsatellite high instability
  • MSI-L microsatellite low instability
  • MSS microsatellite stability
  • Encorafenib is an orally available, highly selective ATP-competitive small molecule RAF kinase inhibitor, which suppresses the RAF/MEK/ERK pathway in tumor cells expressing BRAF V600E mutations, including melanoma and colorectal cancer cell lines.
  • the compound encorafenib which is described by the chemical names “methyl N- ⁇ (2S)-1 -[(4- ⁇ 3-[5-chloro-2- fluoro-3-(methanesulfonamido)phenyl]-1 -(propan-2-yl)-1 H-pyrazol-4-yl ⁇ pyrimidin-2- yl)amino]propan-2-yl ⁇ carbamate” and “methyl N-[(2S)-1 -( ⁇ 4-[3-(5-chloro-2-fluoro-3- methanesulfonamidophenyl)-1 -(propan-2-yl)-1 H-pyrazol-4-yl]pyrimidin-2-yl ⁇ amino)propan-2- yl]carbamate” (and is also referred to as “PF-07263896”, “LGX818”, and “ONO-7702”) has the structure:
  • Encorafenib, and pharmaceutically acceptable salts thereof, and methods of treating cancer using encorafenib are disclosed in International Publication No. WO 2011/025927 published March 3, 201 1. Additional methods of treating cancer using encorafenib, and pharmaceutically acceptable salts thereof, are disclosed in International Publication Nos. WO 2013/070996 published May 16, 2013, WO 2014/018725 published January 30, 2014, WO 2014/025688 published February 13, 2014, WO 2014/072493 published May 15, 2014, WO 2014/147573 published September 25, 2014, and WO 2017/210538 published December 7, 2017.
  • BRAFTOVI® (encorafenib) is indicated, in combination with binimetinib, for the treatment of patients with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation (BRAFTOVI® (encorafenib), United States Prescribing Information, 2018, Colorado, Array BioPharma Inc., a wholly owned subsidiary of Pfizer Inc., the contents of which are incorporated herein by reference in their entirety).
  • BRAFTOVI® (encorafenib) also indicated in combination with cetuximab for the treatment of adult patients with metastatic colorectal cancer (mCRC) with a BRAF V600E mutation, after prior therapy (BRAFTOVI® (encorafenib), United States Prescribing Information, 2020, Colorado, Array BioPharma Inc., a wholly owned subsidiary of Pfizer Inc., the contents of which are incorporated herein by reference in their entirety).
  • Erbitux® an epidermal growth factor receptor (EGFR) antagonist
  • EGFR epidermal growth factor receptor
  • Cetuximab an epidermal growth factor receptor antagonist
  • PD-1 is recognized as an important molecule in immune regulation and the maintenance of peripheral tolerance. PD-1 is moderately expressed on naive T, B and NKT cells and up- regulated by T/B cell receptor signalling on lymphocytes, monocytes and myeloid cells (Sharpe, Arlene H et aL, The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nature Immunology (2007); 8:239-245).
  • PD-L1 Two known ligands for PD-1 , PD-L1 (B7-H1 ) and PD-L2 (B7-DC), are expressed in human cancers arising in various tissues.
  • PD-L1 expression correlated with poor prognosis and reduced overall survival irrespective of subsequent treatment (Dong, Haidong et aL, Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med.
  • Neoplasia (2006) 8: 190-198; Hamanishi, Junzo et aL, Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer.
  • PD-1 expression on tumor infiltrating lymphocytes was found to mark dysfunctional T cells in breast cancer and melanoma (Ghebeh, Hazem et aL, Foxp3+ tregs and B7-H1 +/PD-1 + T lymphocytes co-infiltrate the tumor tissues of high-risk breast cancer patients: implication for immunotherapy.
  • Pembrolizumab (KEYTRUDA®, Merck & Co., Inc., Rahway, NJ, USA) is a potent humanized immunoglobulin G4 (lgG4) mAb with high specificity of binding to the programmed cell death 1 (PD-1) receptor, thus inhibiting its interaction with programmed cell death ligand 1 (PD-L1) and programmed cell death ligand 2 (PD-L2). Based on preclinical in vitro data, pembrolizumab has high affinity and potent receptor blocking activity for PD-1 .
  • Pembrolizumab Keytruda® (pembrolizumab) is indicated for the treatment of patients across a number of indications and is indicated for the first-line treatment of patients with unresectable or metastatic CRC that is microsatellite instability-high or mismatch repair deficient (MSI-H/dMMR).
  • Pembrolizumab is the current standard of care for first line MSI-H/dMMR mCRC.
  • This invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with the PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with the PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging overall survival as compared to treatment with the PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging overall survival as compared to treatment with the PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging overall survival as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging overall survival as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging objective response as compared to treatment with PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging objective response as compared to treatment with PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging objective response as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging objective response as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging duration of response as compared to treatment with the PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging duration of response as compared to treatment with the PD-1 antagonist alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging duration of response as compared to treatment with pembrolizumab alone.
  • This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging duration of response as compared to treatment with pembrolizumab alone.
  • the encorafenib or a pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 150 mg or about 225 mg or about 300 mg calculated as the free base equivalent.
  • the encorafenib or a pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 300 mg calculated as the free base equivalent.
  • the encorafenib is a free base.
  • the encorafenib is formulated for oral administration. In one embodiment, the encorafenib is formulated as a capsule. In one emboidment, the capsule formulation of encorafenib comprises 50 mg of encorafenib as a free base. In one emboidment, the capsule formulation of encorafenib comprises 75 mg of encorafenib as a free base.
  • the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 300 mg/m 2 or about 400 mg/m 2 or about 500 mg/m 2 .
  • the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 500 mg/m 2 .
  • the pembrolizumab is administered as an intravenous infusion every 6 weeks at a dose of about 400 mg. In one embodiment of the present invention, the pembrolizumab is administered as an intravenous infusion every 3 weeks at a dose of about 200 mg.
  • the pembrolizumab is administered as an intravenous infusion every three weeks at a dose of about 2 mg/kg.
  • the colorectal cancer is metastatic or unresectable colorectal cancer.
  • the colorectal cancer is not a colorectal cancer which is RAS -mutant or for which RAS mubation status is unknown.
  • the subject is a human. In one embodiment, the subject is an adult patient. In one embodiment subject is a pediatric patient.
  • a numerically defined parameter e.g., the dose of a encorafenib, or a pharmaceutically acceptable salt thereof, the dose of a a BRAF inhibitor and the like
  • the parameter may vary by as much as 10% above or below the stated numerical value for that parameter.
  • a dose of about 1 .0 mg once daily should be understood to mean that the dose may vary between 0.9 mg once daily and 1 .1 mg once daily.
  • MSI-H/dMMR refers to a cancer that is microsatellite instability-high (MSI-H), mismatch repair deficient (dMMR), or both MSI-H and dMMR.
  • BRAF V600E-mutant, MSI-H/dMMR refers to a cancer that has a BRAF V600E mutation and is also MSI-H, dMMR, or MSI-H and dMMR.
  • Abnormal cell growth refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). Abnormal cell growth may be benign (not cancerous), or malignant (cancerous).
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth.
  • cancer refers to solid tumors.
  • the term “cancer” includes, but is not limited to, a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of different type from latter one.
  • An example of cancer for purposes of the present invention includes metastatic colorectal cancer.
  • metalstatic also known as “secondary cancer” as used herein refers to a type of cancer that originates in one tissue type, but then spreads to one or more tissues outside of the (primary) cancer’s origin.
  • a “patient” or “subject” refers to any single subject for which therapy is desired or that is participating in a clinical trial, epidemiological study or used as a control, including humans and mammalian veterinary patients such as cattle, horses, dogs and cats. In certain preferred embodiments, the subject is a human.
  • a “patient” or “subject” according to the combination of this invention may have BRAF V600E mutant, MSI-H/dMMR colorectal cancer, i.e., a colorectal cancer that: 1 ) has a BRAF V600E mutation and is dMMR; 2) has a BRAF V600E mutation and is MSI-H; or 3) has a BRAF V600E mutation and is dMMR and MSI-H.
  • a BRAF V600E mutation may be detected, for example, by analyzing a biopsy of the cancer or a peripheral blood sample (i.e., liquid biopsy) (e.g., ctDNA genetic testing) using DNA-sequencing (i.e., polymerase chain reaction (PCR)) or next generation sequencing (NGS).
  • dMMR status may be identified, for example, by analyzing a biopsy of the cancer.
  • MSI-H status may be identified, for example, by analyzing a biopsy of the cancer or a peripheral blood sample (i.e., liquid biopsy) (e.g., ctDNA genetic testing).
  • dMMR and/or MSI status is identified by a protein-based assay (such as by an immunoassay, such as an enzyme-linked immunosorbent assay (ELISA) or immunohistochemistry (IHC)), PCR assay (such as a real-time reverse transcriptase PCR assay) or NGS.
  • a protein-based assay such as by an immunoassay, such as an enzyme-linked immunosorbent assay (ELISA) or immunohistochemistry (IHC)
  • PCR assay such as a real-time reverse transcriptase PCR assay
  • treat or “treating” a cancer as used herein means to administer a combination therapy according to the present invention to a subject having cancer, or diagnosed with cancer, to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastases or tumor growth, reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of treating as "treating” is defined immediately above.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cell; inhibiting metastasis or neoplastic cells; shrinking or decreasing the size of tumor; remission of the cancer; decreasing symptoms resulting from the cancer; increasing the quality of life of the subject; delaying the progression the cancer; curing the cancer; overcoming one or more resistance mechanisms of the cancer; and / or prolonging survival of the subject.
  • Positive therapeutic effects in cancer can be measured in a number of other ways (see, for example, W. A. Weber, J. Nucl. Med. 50:1 S-10S (200)).
  • the treatment achieved by a combination of the invention is any of the partial response (PR), complete response (CR), overall response (OR), objective response rate (ORR), progression free survival (PFS), and overall survival (OS).
  • response to a combination of the invention is any of PFS, OR or OS.
  • PFS refers to the length of time during and after the treatment of cancer, that a patient lives with the cancer but it does not get worse (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “PFS” refers the time from randomization to the date of the first documentation of progressive disease (PD) per RECIST v1 .1 or death due to any cause, whichever occurs first. In one embodiment, “PFS” refers to the length of time during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein, that a patient lives with the cancer but it does not get worse.
  • PFS refers to the length of time during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein, that a patient lives with the cancer but it does not get worse as compared to treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer with pembrolizumab alone.
  • OS refers to a prolongation in life expectancy as compared to untreated subjects (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “OS” refers to the time from the date of randomization to the date of death due to any cause. In one embodiment, “OS” refers to the prolongation of life expectancy during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein.
  • OS refers to the prolongation of life expectancy during and after the treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone.
  • OR refers to confirmed complete response (CR) or confirmed PR (partial response), based on investigator assessment per RECIST v1 .1 , from the date of randomization until the date of the first documentation of progressive disease (PD), death or start of new anticancer therapy.
  • “OR” refers to confirmed complete response (CR) or confirmed PR (partial response) during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone.
  • ORR as used herein is defined as the percentage of people in a study or treatment group who have a partial or complete response to the treatment within a certain period of time (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “ORR” refers to the percentage of people in a study or treatment group who have a partial or complete response to the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone.
  • CR refers to the disappearance of all signs of cancer (e.g., tumor lesions) in the body in response to treatment (also referred to as complete remission) (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “CR” refers to the disappearance of all signs of cancer (e.g., tumor lesions) in the body in response to the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer with pembrolizumab alone.
  • PR refers to a decrease in the size of a tumor as determined per RECIST v1 .1 , or in the extent of cancer in the body, in response to treatment (also referred to as partial remission) (National Cancer Institute, Dictionary of Cancer Terms).
  • PR refers to a decrease in the size of a tumor as determined per RECIST v1 .1 , or in the extent of cancer in the body, in response to treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone.
  • PD as used herein refers to a cancer that is growing, spreading, or getting worse (National Cancer Institute, Dictionary of Cancer Terms).
  • Response to a combination of the invention may be assessed using Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1 ) response criteria.
  • the treatment regimen for a combination of the invention that is effective to treat a subject in need thereof may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the subject.
  • any of the aspects of the invention may not be effective in achieving a positive therapeutic effect in every subject, it should do so in a statistically significant number of subjects as determined by any statistical test known in the art such as, but not limited to, the Cox log-rank test, the Cochran- Mantel-Haenszel log-rank test, the Student’s t-test, the chi2-test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstrat-test and the Wilcon on-test.
  • any statistical test known in the art such as, but not limited to, the Cox log-rank test, the Cochran- Mantel-Haenszel log-rank test, the Student’s t-test, the chi2-test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstrat-test and the Wilcon on-test.
  • treatment regimen and “dosing regimen” are used interchangeably to refer to the dose and timing of administration of each therapeutic agent in a combination of the invention.
  • “Ameliorating” means a lessening or improvement of one or more symptoms as compared to not administering a therapeutic agent of a method or regimen of the invention. “Ameliorating” also includes shortening or reduction in duration of a symptom.
  • an “effective dosage,” “effective amount,” “therapeutically effective amount,” or “therapeutically effective dosage” of a drug, therapeutic agent, or pharmaceutical composition is an amount to affect any one or more beneficial or desired, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • an effective amount refers to that amount which has the effect of (1 ) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, (4) relieving to some extent (or, preferably, eliminating) one or more signs or symptoms associated with the cancer, (5) enhancing the effect of another medication, and / or (6) delaying the progression of the disease of the subject.
  • an effective amount refers to that amount which has the effect of prolonging one or more of progression-free survival, overall survival, objective response, and/or duration of response.
  • An effective dosage can be administered in one or more administrations.
  • dosage of a therapeutically effective amount of a compound or pharmaceutical composition thereof may be achieved in conjunction with another therapy.
  • a therapeutically effective amount may be considered in the context of administering one or more therapies (e.g., one or more anticancer agents), and a single agent may be considered to be given in a therapeutically effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • Tumor as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size, and includes primary tumors and secondary neoplasms.
  • a solid tumor is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Examples of solid tumors are sarcomas, carcinomas, and lymphomas. Leukaemia’s (cancers of the blood) generally do not form solid tumors (National Cancer Institute, Dictionary of Cancer Terms).
  • tumor size refers to the total size of the tumor which can be measured as the length and width of a tumor.
  • Tumor size may be determined by a variety of methods known in the art, such as, e.g., by measuring the dimensions of tumor(s) upon removal from the subject, e.g., using callipers, or while in the body using imaging techniques, e.g., ultrasound, a computed tomography (CT) scan (also referred to as a CAT scan), or magnetic resonance imaging (MRI).
  • CT computed tomography
  • MRI magnetic resonance imaging
  • the term “previously untreated” means the subject has not received prior systemic regimens for metastatic CRC.
  • “previously untreated” does not include subjects with early-stage cancer (e.g., Stages l-lll) treated with surgery followed by chemotherapy (e.g., treatment in the adjuvant setting) or who have received prior systemic neoadjuvant therapy with or without radiation who present with new lesions or evidence of recurrence of the cancer during or within 6 months of the last dose of chemotherapy.
  • systemic therapy means treatment using substances that travel through the bloodstream, reaching and affecting cells all over the body (National Cancer Institute, Dictionary of Cancer Terms).
  • systemic regimens i.e., therapies
  • BRAF inhibitors e.g., encorafenib, dabrafenib, vemurafenib, or XL281/BMS-908662
  • EGFR inhibitors e.g., cetuximab or panitumumab
  • immune checkpoint inhibitors such as PD-1 antagonists, or agents directed to another stimulatory or co-inhibitory T-cell receptor (e.g., CTLA-4, 0X40, or CD137).
  • neoadjuvant means a treatment given as a first step to shrink a tumor before the main treatment (e.g., a combination therapy or the present invention).
  • main treatment e.g., a combination therapy or the present invention.
  • neoadjuvant treatment include chemotherapy, radiation therapy and hormone therapy (National Cancer Institute, Dictionary of Cancer Terms).
  • antibody refers to any form of antibody that exhibits the desired biological or binding activity. Thus, it is used in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized, fully human antibodies, chimeric antibodies and camelized single domain antibodies.
  • Monoclonal antibodies including full length monoclonal antibodies
  • polyclonal antibodies include multispecific antibodies (e.g., bispecific antibodies), humanized, fully human antibodies, chimeric antibodies and camelized single domain antibodies.
  • Parental antibodies are antibodies obtained by exposure of an immune system to an antigen prior to modification of the antibodies for an intended use, such as humanization of an antibody for use as a human therapeutic.
  • the basic antibody structural unit comprises a tetramer.
  • Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function.
  • human light chains are classified as kappa and lambda light chains.
  • human heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids. See generally, Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989).
  • variable regions of each light/heavy chain pair form the antibody binding site.
  • an intact antibody has two binding sites.
  • the two binding sites are, in general, the same.
  • variable domains of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • the CDRs are usually aligned by the framework regions, enabling binding to a specific epitope.
  • both light and heavy chains variable domains comprise FR1 , CDR1 , FR2, CDR2, FR3, CDR3 and FR4.
  • the assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al.; National Institutes of Health, Bethesda, Md. ; 5 th ed.; NIH Publ. No.
  • antibody fragment or “antigen binding fragment” refers to antigen binding fragments of an antibody, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full-length antibody, e.g. fragments that retain one or more CDR regions, e.g. all six CDRs.
  • antibody binding fragments include, but are not limited to, Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and multispecific antibodies formed from antibody fragments.
  • An antibody that “specifically binds to” a specified target protein is an antibody that exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity.
  • An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g. without producing undesired results such as false positives.
  • Antibodies, or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20- times greater, and most preferably at least 100-times greater than the affinity with non-target proteins.
  • an antibody is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human PD-1 or human PD-L1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • CDR or “CDRs” as used herein means complementarity determining region(s) in a immunoglobulin variable region, defined using the Kabat numbering system, unless otherwise indicated.
  • Constantly modified variants or “conservative substitution” refers to substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g. charge, sidechain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity or other desired property of the protein, such as antigen affinity and/or specificity.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)).
  • substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity. Exemplary conservative substitutions are set forth in Table 1 below.
  • Framework region or “FR” as used herein means the immunoglobulin variable regions excluding the CDR regions.
  • Kabat as used herein means an immunoglobulin alignment and numbering system pioneered by Elvin A. Kabat ((1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.).
  • conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731 .
  • PD-1 antagonist means any chemical compound or biological molecule that blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or Natural Killer T cell) and in specific embodiments also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1 .
  • Alternative names or synonyms for PD-1 and its ligands include: PDCD1 , PD1 , CD279 and SLEB2 for PD-1 ; PDCD1 L1 , PDL1 , B7H1 , B7-4, CD274 and B7-H for PD-L1 ; and PDCD1 L2, PDL2, B7-DC, Btdc and CD273 for PD-L2.
  • the PD-1 antagonist blocks binding of human PD-L1 to human PD-1 , and in specific embodiments blocks binding of both human PD-L1 and PD-L2 to human PD-1.
  • Human PD-1 amino acid sequences can be found in NCBI Locus No.: NP 005009.
  • Human PD- L1 and PD-L2 amino acid sequences can be found in NCBI Locus No.: NP 054862 and NP 079515, respectively.
  • “Pembrolizumab” (formerly known as MK-3475, SCH 900475 and lambrolizumab) alternatively referred to herein as “pembro,” is a humanized lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 2, pages 161-162 (2013) and which comprises the heavy and light chain amino acid sequences and CDRs described in Table 2.
  • Pembrolizumab has been approved by the U.S. FDA as described in the Prescribing Information for KEYTRUDATM (Merck & Co., Inc., Rahway, NJ, USA; initial U.S. approval 2014, updated March 2021).
  • a “pembrolizumab variant” or “a variant thereof” pertaining to a pembrolizumab sequence means a monoclonal antibody that comprises heavy chain and light chain sequences that are substantially identical to those in pembrolizumab, except for having three, two or one conservative amino acid substitutions at positions that are located outside of the light chain CDRs and six, five, four, three, two or one conservative amino acid substitutions that are located outside of the heavy chain CDRs, e.g., the variant positions are located in the FR regions or the constant region, and optionally has a deletion of the C-terminal lysine residue of the heavy chain.
  • pembrolizumab and a pembrolizumab variant comprise identical CDR sequences, but differ from each other due to having a conservative amino acid substitution at no more than three or six other positions in their full-length light and heavy chain sequences, respectively.
  • a pembrolizumab variant is substantially the same as pembrolizumab with respect to the following properties: binding affinity to PD-1 and ability to block the binding of each of PD-L1 and PD-L2 to PD-1.
  • the methods and combination therapies of the present invention are useful for treating cancer.
  • the cancer is a previously untreated BRAF V600E mutant, MSI- H/dMMR colorectal cancer.
  • Colorectal cancer is also referred to as colorectal adenocarcinoma.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • the BRAF inhibitor is selected from encorafenib, dabrafenib, vemurafenib, N-[3-(5-chloro-1 H-pyrrolo[2,3-b]pyridin-3-ylcarbonyl)-2,4-difluorophenyl]propane-
  • WO 2020/261 156 A1 including, for example, a compound selected from N-(3-((3,5-dimethyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)-2,4-difluorophenyl)propane-1 -sulfonamide, N-(2-chloro-3-((3,5- dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-3-fluoropropane-1 -sulfonamide, N-(2- chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4,5-difluorophenyl)propane-1 - sulfonamide, N-(2-chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl
  • WO 2021/250521 published December 16, 2021 , including, for example, N-(2-chloro-4-fluoro-3-((5-fluoro-3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-2-azabicyclo[2.1 ,1]hexane-2-sulfonamide, (R)-N-(2- chloro-4-fluoro-3-((5-fluoro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-3- fluoropyrrolidine-1 -sulfonamide, and N-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3-fluoroazetidine-1 -sulfonamide, and pharmaceutically acceptable salt thereof.
  • the BRAF inhibitor is selected from encorafenib or a pharmaceutically acceptable salt thereof, /V-(2-chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4- fluorophenyl)-3-fluoropropane-1 -sulfonamide or a pharmaceutically acceptable salt thereof, and N-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3- fluoroazetidine-1 -sulfonamide or a pharmaceutically acceptable salt thereof.
  • the BRAF inhibitor is encorafenib or a pharmaceutically acceptable salt thereof.
  • the BRAF inhibitor is encorafenib as a free base.
  • the EGFR inhibitor is selected from cetuximab (Erbitux®), panitumumab (Vectibix®), osimertinib (Tagrisso®), erlotinib (Tarceva®), gefitinib (Iressa®), necitumumab (PortrazzaTM), neratinib (Nerlynx®), lapatinib (Tykerb®), vandetanib (Caprelsa®), brigatinib (Alunbrig®).
  • the EGFR inhibitor is cetuximab.
  • the PD-1 antagonist useful in the treatment, medicaments and uses of the present invention include a monoclonal antibody (mAb), or antigen binding fragment thereof, that specifically binds to PD-1 or PD-L1 , and preferably specifically binds to human PD- 1 or human PD-L1 .
  • the mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region.
  • the human constant region is selected from the group consisting of IgG 1 , lgG2, lgG3 and lgG4 constant regions, and in some embodiments, the human constant region is an IgG 1 or lgG4 constant region.
  • the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab') 2 , scFv and Fv fragments.
  • Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include: pembrolizumab (also known as MK-3475), a humanized lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 2, pages 161-162 (2013) and that comprises the heavy and light chain amino acid sequences shown in Table 2; nivolumab (BMS-936558), a human lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No.
  • mAbs that bind to human PD-L1 are described in US8383796.
  • Specific antihuman PD-L1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include BMS-936559, MEDI4736, and MSB0010718C.
  • the PD-1 antagonist is pembrolizumab (KEYTRUDATM, Merck & Co., Inc., Rahway, NJ, USA), nivolumab (OPDIVOTM, Bristol-Myers Squibb Company, Princeton, NJ, USA), atezolizumab (TECENTRIQTM, Genentech, San Francisco, CA, USA), durvalumab (IMFINZITM, AstraZeneca Pharmaceuticals LP, Wilmington, DE), cemiplimab (LIBTAYOTM, Regeneron Pharmaceuticals, Tarrytown, NY, USA) avelumab (BAVENCIOTM, Merck KGaA, Darmstadt, Germany) or dostarlimab (JEMPERLITM, GlaxoSmithKline LLC, Philadelphia, PA).
  • pembrolizumab KYTRUDATM, Merck & Co., Inc., Rahway, NJ, USA
  • OPDIVOTM Bristol-Myers Squibb Company, Princeton, NJ, USA
  • the PD-1 antagonist is pidilizumab (U.S. Pat. No. 7,332,582), AMP-514 (Medlmmune LLC, Gaithersburg, MD, USA), PDR001 (U.S. Pat. No. 9,683,048), BGB-A317 (U.S. Pat. No. 8,735,553), or MGA012 (MacroGenics, Rockville, MD).
  • the PD-1 antagonist useful in the methods of the invention is an anti-PD-1 antibody that blocks the binding of PD-1 to PD-L1 and PD-L2.
  • the PD-1 antagonist is a monoclonal antibody, or antigen binding fragment thereof, that comprises: (a) a light chain variable region comprising light chain CDR1 , CDR2 and CDR3 of SEQ ID NOs: 1 , 2 and 3, respectively and (b) a heavy chain variable region comprising heavy chain CDR1 , CDR2 and CDR3 of SEQ ID NOs: 6, 7 and 8, respectively.
  • the PD-1 antagonist is a monoclonal antibody, or antigen binding fragment thereof, that specifically binds to human PD-1 and comprises (a) a heavy chain variable region comprising SEQ ID NO:9 or a variant thereof, and (b) a light chain variable region comprising SEQ ID NO:4 or a variant thereof.
  • a variant of a heavy chain variable region sequence is identical to the reference sequence except having up to six conservative amino acid substitutions in the framework region (i.e. , outside of the CDRs).
  • a variant of a light chain variable region sequence is identical to the reference sequence except having up to three conservative amino acid substitutions in the framework region (i.e., outside of the CDRs).
  • the PD-1 antagonist is a monoclonal antibody that specifically binds to human PD-1 and comprises (a) a heavy chain comprising SEQ ID NO: 10 and (b) a light chain comprising SEQ ID NO:5.
  • the PD-1 antagonist is an anti-PD-1 antibody that comprises two heavy chains and two light chains, and wherein the heavy and light chains comprise the amino acid sequences in SEQ ID NQ:10 and SEQ ID NO:5, respectively.
  • the PD-1 antagonist inhibits the binding of PD-L1 to PD-1 , and in specific embodiments also inhibits the binding of PD-L2 to PD-1 .
  • the PD-1 antagonist is a monoclonal antibody, or an antigen binding fragment thereof, that specifically binds to PD-1 or to PD-L1 and blocks the binding of PD-L1 to PD-1 .
  • Table 2 below provides a list of the amino acid sequences of exemplary anti-PD-1 mAbs for use in the treatment method, medicaments and uses of the present invention.
  • Table 2. Exemplary PD-1 Antibody Sequences Table 3. Additional PD-1 Antibodies and Antigen Binding Fragments Useful in the Formulations, Methods and Uses of the Invention.
  • the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain constant region, e.g. a human constant region, such as g1 , g2, g3, or g4 human heavy chain constant region or a variant thereof.
  • the anti- PD-1 antibody or antigen-binding fragment thereof comprises a light chain constant region, e.g. a human light chain constant region, such as lambda or kappa human light chain region or a variant thereof.
  • the human heavy chain constant region can be g4 and the human light chain constant region can be kappa.
  • the Fc region of the antibody is g4 with a Ser228Pro mutation (Schuurman, J et. al., Mol. Immunol. 38: 1 -8, 2001 ).
  • different constant domains may be appended to humanized VL and VH regions derived from the CDRs provided herein. For example, if a particular intended use of an antibody (or fragment) of the present invention were to call for altered effector functions, a heavy chain constant domain other than human IgG 1 may be used, or hybrid lgG1/lgG4 may be utilized.
  • human lgG1 antibodies provide for long half-life and for effector functions, such as complement activation and antibody-dependent cellular cytotoxicity, such activities may not be desirable for all uses of the antibody.
  • a human lgG4 constant domain for example, may be used.
  • the present invention includes the use of anti-PD-1 antibodies or antigen-binding fragments thereof which comprise an lgG4 constant domain.
  • the lgG4 constant domain can differ from the native human lgG4 constant domain (Swiss-Prot Accession No.
  • the PD-1 antagonist is an antibody or antigen binding protein that has a variable light domain and/or a variable heavy domain with at least 95%, 90%, 85%, 80%, 75% or 50% sequence identity to one of the variable light domains or variable heavy domains described above, and exhibits specific binding to PD-1 .
  • the PD-1 antagonist is an antibody or antigen binding protein comprising variable light and variable heavy domains having up to 1 , 2, 3, 4, or 5 or more amino acid substitutions, and exhibits specific binding to PD-1
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
  • this invention relates to a BRAF inhibitor for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the BRAF inhibitor is used in combination with an EGFR inhibitor and a PD-1 antagonist.
  • this invention relates to an EGFR inhibitor for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the EGFR is used in combination with a BRAF inhibitor and a PD-1 antagonist.
  • this invention relates to a PD-1 antagonist for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the PD-1 antagonist is used in combination with a BRAF inhibitor and an EGFR inhibitor.
  • this invention relates to encorafenib, or a pharmaceutically acceptable salt thereof, for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the encorafenib, or a pharmaceutically acceptable salt thereof, is used in combination with cetuximab and pembrolizumab.
  • this invention relates to cetuximab for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the cetuximab is used in combination with encorafenib, or a pharmaceutically acceptable salt thereof, and pembrolizumab.
  • this invention relates to pembrolizumab for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the pembrolizumab is used in combination with encorafenib, or a pharmaceutically acceptable salt thereof, and cetuximab.
  • this invention relates to the use of encorafenib, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the medicament used in combination with cetuximab and pembrolizumab.
  • this invention relates to the use of cetuximab in the manufacture of a medicament for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the medicament used in combination with encorafenib or a pharmaceutically acceptable salt thereof, and pembrolizumab.
  • this invention relates to the use of pembrolizumab in the manufacture of a medicament for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the medicament used in combination with encorafenib or a pharmaceutically acceptable salt thereof, and cetuximab.
  • this invention relates to a pharmaceutical composition comprising encorafenib, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient, for use in the treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer in a subject in need thereof, wherein the pharmaceutical composition comprising the encorafenib, or a pharmaceutically acceptable salt thereof, is used in combination with a pharmaceutical composition comprising cetuximab and at least one pharmaceutically acceptable excipient, and a pharmaceutical composition comprising pembrolizumab and at least one pharmaceutically acceptable excipient.
  • this invention relates to a pharmaceutical composition comprising cetuximab and at least one pharmaceutically acceptable excipient, for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the pharmaceutical composition comprising the cetuximab is used in combination with a pharmaceutical composition comprising encorafenib or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient, and a pharmaceutical composition comprising pembrolizumab and at least one pharmaceutically acceptable excipient.
  • this invention relates to a pharmaceutical composition comprising pembrolizumab and at least one pharmaceutically acceptable excipient for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the pharmaceutical composition comprising the pembrolizumab is used in combination with a pharmaceutical composition comprising encorafenib or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient, and a pharmaceutical composition comprising cetuximab and at least one pharmaceutically acceptable excipient.
  • the colorectal cancer is BRAF V600E-mutant, MSI-H colorectal cancer.
  • the colorectal cancer is BRAF V600E-mutant, dMMR colorectal cancer.
  • the colorectal cancer is BRAF V600E-mutant, MSI-H, dMMR colorectal cancer.
  • the colorectal cancer is metastatic or unresectable colorectal cancer. In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, progression-free survival is prolonged as compared to treatment with pembrolizumab alone.
  • objective response is prolonged as compared to treatment with pembrolizumab alone.
  • duration of response is prolonged as compared to treatment with pembrolizumab alone.
  • the encorafenib is the free base.
  • the subject is a mammal.
  • the subject is a human.
  • the BRAF V600E mutation is detected in tumor tissue by PCR or NGS.
  • dMMR status is identified by IHC.
  • MSI-H status is identified by DNA-based testing (PCR).
  • Each therapeutic agent of the methods and combination therapies of the present invention may be administered either alone, or in a medicament (also referred to herein as a pharmaceutical composition) comprising the therapeutic agent and at least one pharmaceutically acceptable excipient, according to pharmaceutical practice.
  • the term “combination therapy” refers to the administration of each therapeutic agent of the combination therapy of the invention, either alone or in a medicament, either sequentially or concurrently, wherein the agents are dosed in amounts which together are effective in treating the cancer.
  • the term “sequential” or “sequentially” refers to the administration of each therapeutic agent of the combination therapy of the invention, either alone or in a medicament, one after the other, wherein each therapeutic agent can be administered in any order and with variable intervening time limits.
  • Sequential administration is particularly useful when the therapeutic agents in the combination therapy are in different dosage forms, for example, one agent is a tablet and the second agent and/or third agent is a sterile liquid, and/or the therapeutic agents are administered by different routes of administration, for example one agent is administered orally and the second agent and/or third agent is administered intravenously, and/or the therapeutic agents are administered according to different dosing schedules, for example, one agent is administered daily, and the second agent and/or third agent is administered less frequently such as weekly, every two weeks, or every 6 weeks.
  • the term “concurrently” refers to the administration of each therapeutic agent in the combination therapy of the invention, either alone or in separate medicaments, wherein the second and third therapeutic agents are administered immediately after the first therapeutic agent, but that the therapeutic agents can be administered in any order.
  • the encorafenib is formulated for oral administration. In one embodiment, the encorafenib is formulated as a capsule. In one emboidment, the capsule formulation of encorafenib comprises 50 mg of encorafenib as a free base. In one emboidment, the capsule formulation of encorafenib comprises 75 mg of encorafenib as a free base.
  • an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof is administered orally as a once daily (QD) dosage of about 150 mg or about 225 mg or about 300 mg calculated as the free base equivalent. In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent. In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent. In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent.
  • an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m 2 or about 400 mg/m 2 or about 500 mg/m 2 . In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m 2 . In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m 2 . In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m 2 . In one embodiment, cetuximab is administered as a 120-minute intravenous infusion. In one embodiment, the subject is administered a premedication (e.g., antihistamine or antipyretic) prior to administration of cetuximab.
  • a premedication e.g
  • an effective dosage of pembrolizumab is administered at a dose of about 400 mg every 6 weeks (Q6W). In one embodiment, an effective dosage of pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W). In one embodiment, pembrolizumab is administered as a 30-minute (-5 minutes /+10 minutes) intravenous infusion. In one embodiment, the selected dose of pembrolizumab is administered by IV infusion over a time period of between 25 and 40 minutes, or about 30 minutes. In one embodiment, the subject is administered a premedication (e.g., antihistamine or antipyretic) prior to administration of pembrolizumab. In one embodiment, the selected dose of pembrolizumab is administered by subcutaneous injection.
  • a premedication e.g., antihistamine or antipyretic
  • an effective dosage of pembrolizumab is administered as an intravenous infusion at a dose of about 200 mg every 3 weeks (Q3W).
  • the method comprises administering 2 mg/kg of pembrolizumab to the patient about every three weeks.
  • the patient is a pediatric patient.
  • At least one of the therapeutic agents in the combination therapy is administered using the same dosage regimen (dose, frequency, and duration of treatment) that is typically employed when the agent is used as a monotherapy for treating the same cancer.
  • the subject received a lower total amount of at least one of the therapeutic agents in the combination therapy than when the same agent is used as a monotherapy, for example a lower dose of therapeutic agent, a reduced frequency of dosing and / or a shorter duration of dosing.
  • encorafenib is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • encorafenib is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent
  • cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m 2
  • pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
  • cetuximab and pembrolizumab are administered on the same day, the encorafenib or a pharmaceutically acceptable salt thereof is administered prior to administration of the pembrolizumab, which is then followed by administration of the cetuximab.
  • the cetuximab and the pembrolizumab are administered on the same day, the cetuximab is administered about 30-60 minutes after administration of the pembrolizumab.
  • Dosage regimens may be adjusted to provide the optimum desired response.
  • a therapeutic agent of the combination therapy of the present invention may be administered as a single bolus, as several divided doses administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be particularly advantageous to formulate a therapeutic agent in a dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical excipient.
  • the specification for the dosage unit forms of the invention may be dictated by and directly dependent on (a) the unique characteristics of the therapeutic agent and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • the dose and dosing regimen is adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose may be readily established, and the effective amount providing a detectable therapeutic benefit to a subject may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the subject. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a subject in practicing the present invention.
  • dosage values may vary with the type and severity of the condition to be alleviated and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, taking into consideration factors such as the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
  • the dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values.
  • the present invention encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regimens for administration of the therapeutic agent are well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
  • a “continuous dosing schedule” as used herein is an administration or dosing regimen without dose interruptions, e.g., without days off treatment. Repetition of 21 - or 28-day treatment cycles without dose interruptions between the treatment cycles is an example of a continuous dosing schedule.
  • the compounds of the combination of the present invention can be administered in a continuous dosing schedule.
  • a “pharmaceutical composition” refers to a mixture of one or more of the compounds of the invention, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof as an active ingredient, and at least one pharmaceutically acceptable excipient.
  • excipient is used herein to describe any ingredient other than the therapeutic agents of the combination therapy.
  • the choice of excipient will to a large extent depend on factors such as the mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • excipient includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, carriers, diluents and the like that are physiologically compatible.
  • excipients include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof, and may include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol, or sorbitol in the composition.
  • excipients also include various organic solvents (such as hydrates and solvates).
  • the pharmaceutical compositions may, if desired, contain additional excipients such as flavorings, binders/binding agents, lubricating agents, disintegrants, sweetening or flavoring agents, coloring matters or dyes, and the like.
  • excipients such as citric acid
  • disintegrants such as starch, alginic acid and certain complex silicates
  • binding agents such as sucrose, gelatin and acacia.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes.
  • Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules.
  • excipients therefore, also include lactose or milk sugar and high molecular weight polyethylene glycols.
  • aqueous suspensions or elixirs are desired for oral administration of the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with additional excipients such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • excipients also include pharmaceutically acceptable substances such as wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives, or buffers, which enhance the shelf life or effectiveness of the compound.
  • compositions of each of the therapeutic agents of the combination of the invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, capsules, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions e.g., injectable and infusible solutions
  • tablets, capsules, pills, powders, liposomes and suppositories e.g., suppositories.
  • the form depends on the intended mode of administration and therapeutic application.
  • Administration of a composition comprising a therapeutic agent used in the combination of the invention may be oral, parenteral, topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), or pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal),
  • Oral administration of a solid dosage form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount the therapeutic agent.
  • the oral administration may be in a powder or granule form.
  • the oral dosage form is sub-lingual, such as, for example, a lozenge.
  • the therapeutic agent is ordinarily combined with one or more adjuvants.
  • Such capsules or tablets may comprise a controlled release formulation.
  • the dosage forms also may comprise buffering agents or may be prepared with enteric coatings.
  • oral administration may be in a liquid dosage form.
  • Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as one or more of wetting, emulsifying, suspending, flavoring (e.g., sweetening), or perfuming agents.
  • adjuvants such as one or more of wetting, emulsifying, suspending, flavoring (e.g., sweetening), or perfuming agents.
  • the therapeutic agent may be administered in a parenteral dosage form.
  • Parenteral administration includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion.
  • injectable preparations i.e. , sterile injectable aqueous or oleaginous suspensions
  • suitable dispersing, wetting agents, or suspending agents may be formulated according to the known art using one or more of suitable dispersing, wetting agents, or suspending agents.
  • the therapeutic agent may be administered in a topical dosage form.
  • Topical administration includes, for example, dermal and transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration.
  • Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams.
  • a topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages and microemulsions. Liposomes may also be used.
  • Typical excipients include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol.
  • Penetration enhancers may be incorporated - see, for example, B. C. Finnin and T. M. Morgan, J. Pharm. Sci., vol. 88, pp. 955-958, 1999.
  • Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in a suitable excipient.
  • a typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as crossed linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methylcellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • the therapeutic agent may be conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant.
  • Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1 ,1 ,1 ,2- tetrafluoroethane or 1 ,1 ,1 ,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the therapeutic agent may be administered as a rectal dosage form.
  • rectal dosage form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • compositions of the therapeutic agents may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures.
  • effective formulations and administration procedures are well known in the art and are described in standard textbooks.
  • Formulation of drugs is discussed in, for example, Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman et aL, Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe et aL, Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999.
  • Acceptable excipients are nontoxic to subjects at the dosages and concentrations employed, and may comprise one or more of the following: 1 ) buffers such as phosphate, citrate, or other organic acids; 2) salts such as sodium chloride; 3) antioxidants such as ascorbic acid or methionine; 4) preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol; 5) alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, or m-cresol; 6) low molecular weight (less than about 10 residues) polypeptides; 7) proteins such as serum albumin, gelatin, or immunoglobulins; 8) hydrophilic polymers such as polyvinylpyrrolidon
  • Liposome containing a therapeutic agent may be prepared by methods known in the art (See, for example, Chang, H.L; Yeh, M.K.; Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy; Int J Nanomedicine 2012; 7; 49-60). Particularly useful liposomes may be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • PEG-PE PEG-derivatized phosphatidylethanolamine
  • a therapeutic agent may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules
  • sustained-release preparations may be used. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing a therapeutic agent, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate) or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as those used in leuprolide acetate for depot suspension (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3-hydroxybutyric acid.
  • polyesters for example, poly(2- hydroxyethyl-methacrylate) or poly(vinylalcohol)
  • polylactides copolymers of L-glutamic acid and 7 ethyl-L-glutamate
  • Suitable emulsions may be prepared using commercially available fat emulsions, such as a lipid emulsions comprising soybean oil, a fat emulsion for intravenous administration (e.g., comprising safflower oil, soybean oil, egg phosphatides and glycerin in water), emulsions containing soya bean oil and medium-chain triglycerides, and lipid emulsions of cottonseed oil.
  • a lipid emulsions comprising soybean oil
  • a fat emulsion for intravenous administration e.g., comprising safflower oil, soybean oil, egg phosphatides and glycerin in water
  • emulsions containing soya bean oil and medium-chain triglycerides emulsions containing soya bean oil and medium-chain triglycerides
  • lipid emulsions of cottonseed oil such as a lipid emulsions comprising soybean oil, a
  • the active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g., egg phospholipids, soybean phospholipids or soybean lecithin) and water.
  • an oil e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil
  • a phospholipid e.g., egg phospholipids, soybean phospholipids or soybean lecithin
  • Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%.
  • the fat emulsion may comprise fat droplets between 0.1 and 1.0 pm, particularly 0.1 and 0.5 pm, and have a pH in the range of 5.5 to 8.0.
  • the emulsion compositions may be those prepared by mixing a therapeutic agent with a lipid emulsion comprising soybean oil or the components thereof (soybean oil, egg phospholipids, glycerol and water).
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulized by use of gases. Nebulized solutions may be breathed directly from the nebulizing device, or the nebulizing device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • a drug product intermediate is a partly processed material that must undergo further processing steps before it becomes bulk drug product.
  • a therapeutic agent of the invention may be formulated into drug product intermediate DPI containing the active ingredient in a higher free energy form than the crystalline form.
  • One reason to use a DPI is to improve oral absorption characteristics due to low solubility, slow dissolution, improved mass transport through the mucus layer adjacent to the epithelial cells, and in some cases, limitations due to biological barriers such as metabolism and transporters. Other reasons may include improved solid-state stability and downstream manufacturability.
  • the drug product intermediate contains a therapeutic agent isolated and stabilized in the amorphous state (for example, amorphous solid dispersions (ASDs)).
  • ASSDs amorphous solid dispersions
  • ASD Advanced Drug Delivery
  • SDD spray dried dispersions
  • HME melt extrudates
  • co-precipitates amorphous drug nanoparticles
  • nano-adsorbates amorphous solid dispersions
  • amorphous solid dispersions comprise a therapeutic agent and a polymer excipient.
  • Other excipients as well as concentrations of said excipients and the therapeutic agent are well known in the art and are described in standard textbooks. See, for example, “Amorphous Solid Dispersions Theory and Practice” by Navnit Shah et al.
  • the PD-1 antagonist is included in a pharmaceutical composition with a pharmaceutically acceptable carrier or diluent and may include additional pharmaceutically acceptable excipients.
  • the therapeutic agents of the combination therapies of the present invention may conveniently be combined in the form of a kit suitable for coadministration of the therapeutic agents.
  • the present invention relates to a kit comprising a first container, a second container, a third container, and a package insert, wherein the first container comprises at least one dose of encorafenib or a pharmaceutically acceptable salt thereof, the second container comprises at least one dose of cetuximab, the third container comprises at least one dose of pembrolizumab, and the package insert comprises instructions for treating a subject for cancer using the medicaments.
  • the kit may be particularly suitable for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit typically includes directions for administration and may be provided with a memory aid.
  • the kit may further comprise other materials that may be useful in administering the medicaments, such as diluents, filters, IV bags and lines, needles and syringes, and the like.
  • the purpose of this clinical study is to evaluate the safety and efficacy of encorafenib and cetuximab in combination with pembrolizumab compared with pembrolizumab alone in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC.
  • Subject will be randomized in a 1 :1 ratio to Arm A or Arm B. Randomization will be stratified by ECOG (0 vs 1 ). Participants in Arm B are not eligible for crossover to Arm A. Participants will receive either:
  • Study intervention will be administered until progressive disease (PD) per RECIST v1 .1 or until any of the other protocol-defined criteria for discontinuation of study intervention are met, whichever occurs first. In certain circumstances, continuation of treatment beyond PD may be allowed. In both treatment arms, duration of pembrolizumab treatment will not exceed 18 administrations ( ⁇ 24 months); and in Arm A, treatment with encorafenib and cetuximab will continue beyond ⁇ 24 months until PD per RECIST v1 .1 or until any of the other protocol-defined criteria for discontinuation of study intervention are met, whichever occurs first. After discontinuation of all study intervention, participants will be followed for safety, disease status, subsequent anticancer therapy, and survival status until withdrawal of consent/assent, the participant is lost to follow-up, death, or defined end of study, whichever occurs first.
  • PD progressive disease
  • pembrolizumab No dose adjustments for pembrolizumab is recommended. Pembrolizumab will be withheld for treatment-related toxicities as appropriate. Participants should receive appropriate supportive care measures deemed necessary. The dose of pembrolizumab will remain constant at [400 mg Q6W] for each dose level for encorafenib and cetuximab and in each arm.
  • male or female participants age >16 years at the time of informed consent/assent (or the minimum country specific age of consent if >16). In countries or sites where enrollment of adolescents is not permitted (e.g., Germany), male or female participants age >18 years at the time of informed consent.
  • Baseline scan is defined as the last scan prior to the date of randomization. Note: Baseline scans will be required to be available for subsequent submission to a central radiology vendor to be assessed by the BICR.
  • the archival sample should be from the same tumor block that was used for local BRAF V600E mutation and MSI-H/dMMR testing.
  • This tissue block should be obtained from a biopsy or surgery that was performed within 2 years prior to study enrollment. A newly obtained tumor tissue biopsy must be provided prior to randomization for participants unable to provide adequate archival tumor tissue. If a newly obtained biopsy is taken, the biopsy should be taken from a nontarget lesion when possible.
  • Adequate bone marrow function characterized by the following at screening: a. ANC >1.5 x 109/L b. Platelets >100 x 109/L c. Hemoglobin >9.0 g/dL (without blood transfusions 2 weeks prior to randomization)
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • Impaired gastrointestinal function e.g., uncontrolled nausea, vomiting or diarrhea, malabsorption syndrome, small bowel resection
  • disease which may significantly alter the absorption of oral study intervention or recent changes in bowel function suggesting current or impending bowel obstruction.
  • Clinically significant cardiovascular diseases including any of the following: a. History of acute myocardial infarction, acute coronary syndromes (including unstable angina, coronary artery bypass graft, coronary angioplasty or stenting) ⁇ 6 months prior to randomization. b. Congestive heart failure requiring treatment (New York Heart Association Grade > 2). c. Recent history (one year) or presence of clinically significant cardiac arrhythmias (including uncontrolled atrial fibrillation or uncontrolled paroxysmal supraventricular tachycardia). d. History of thromboembolic or cerebrovascular events ⁇ 12 weeks prior to randomization.
  • Examples include transient ischemic attacks, cerebrovascular accidents, hemodynamically significant (i.e., massive or sub-massive) deep vein thrombosis or pulmonary emboli.
  • Participants with either deep vein thrombosis or pulmonary emboli that do not result in hemodynamic instability are allowed to enroll as long as they are on a stable dose of anticoagulants for at least 4 weeks.
  • Participants with thromboembolic events related to indwelling catheters (including PICC lines) or other procedures may be enrolled.
  • Participants with BBB or with an implanted cardiac pacemaker may enroll into the study upon agreement between the investigator and sponsor or designee. f. LQTS.
  • Active HBV is defined as any of the following:
  • Active HCV is defined as:
  • PFS per investigator defined as the time from randomization until PD based on investigator assessment per RECIST v1 .1 or death due to any cause, whichever occurs first.
  • Objective response defined as confirmed CR or confirmed PR based on investigator assessment per RECIST v1 .1 , from the date of randomization until the date of the first documentation of PD, death or start of new anticancer therapy
  • DOR defined as the time from the first response, until PD based on investigator assessment per RECIST v1 .1 or death due to any cause, whichever occurs first

Abstract

This invention relates to combination therapies useful for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer. In particular, the invention relates to a combination therapy comprising or consisting essentially of a BRAF inhibitor, an EGFR inhibitor, and a PD-1 antagonist.

Description

COMBINATION OF A BRAF INHIBITOR, AN EGFR INHIBITOR, AND A PD-1 ANTAGONIST FOR THE TREATMENT OF BRAF V600E-MUTANT, MSI-H/DMMR COLORECTAL CANCER
This application incorporates the contents of a 23.6 kb xml file created on May 25, 2023 and named “SequenceListing.xml,” which is the sequence listing for this application.
Background of the Invention
The present invention relates to combination therapies useful for the treatment of previously untreated BRAF V600E-mutant, microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) colorectal cancer. In particular, the invention relates to a combination therapy comprising a BRAF inhibitor, an EGFR inhibitor, and an antagonist of a Programmed Death 1 protein (PD-1 ). The invention also relates to associated methods of treatment, pharmaceutical compositions, and pharmaceutical uses.
Approximately 20-25% of BRAF V600E-mutant metastatic colorectal cancer (mCRC) tumors are also microsatellite high/ mismatch repair deficient (MSI-H/dMMR) (Venderbosch et al., Clinical Cancer Research, 2014;20(20):5322-5330; Andre et al., N. Engl. J. Med. 2020;383(23):2207-2218). Microsatellite (MS) refers to DNA sequences in tandem repeats in units of a few nucleotides (1 -6 more) in the genome of a cell, also known as short tandem repeats (str). In the event of deficient mismatch repair (dMMR) function, replication errors arising from microsatellites are not corrected and accumulated, resulting in a change in microsatellite length or base composition, called microsatellite instability (MSI), while causing the genome to exhibit a high mutant phenotype. MSI may be divided into 3 classes depending on the degree of extent: microsatellite high instability (MSI-H), microsatellite low instability (MSI-L), microsatellite stability (MSS). For example, microsatellite testing that shows mutations in 30% or more microsatellites is called microsatellite instability-high (National Cancer Institute, Dictionary of Cancer Terms). Tumors that have a dMMR system can develop MSI-H. Patients with BRAF V600E-mutant mCRC that is also MSI-H/dMMR are associated with a poor prognosis.
Encorafenib is an orally available, highly selective ATP-competitive small molecule RAF kinase inhibitor, which suppresses the RAF/MEK/ERK pathway in tumor cells expressing BRAF V600E mutations, including melanoma and colorectal cancer cell lines. The compound encorafenib, which is described by the chemical names “methyl N-{(2S)-1 -[(4-{3-[5-chloro-2- fluoro-3-(methanesulfonamido)phenyl]-1 -(propan-2-yl)-1 H-pyrazol-4-yl}pyrimidin-2- yl)amino]propan-2-yl}carbamate” and “methyl N-[(2S)-1 -({4-[3-(5-chloro-2-fluoro-3- methanesulfonamidophenyl)-1 -(propan-2-yl)-1 H-pyrazol-4-yl]pyrimidin-2-yl}amino)propan-2- yl]carbamate” (and is also referred to as “PF-07263896”, “LGX818”, and “ONO-7702”) has the structure:
Figure imgf000003_0001
Encorafenib
Encorafenib, and pharmaceutically acceptable salts thereof, and methods of treating cancer using encorafenib, are disclosed in International Publication No. WO 2011/025927 published March 3, 201 1. Additional methods of treating cancer using encorafenib, and pharmaceutically acceptable salts thereof, are disclosed in International Publication Nos. WO 2013/070996 published May 16, 2013, WO 2014/018725 published January 30, 2014, WO 2014/025688 published February 13, 2014, WO 2014/072493 published May 15, 2014, WO 2014/147573 published September 25, 2014, and WO 2017/210538 published December 7, 2017.
BRAFTOVI® (encorafenib) is indicated, in combination with binimetinib, for the treatment of patients with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation (BRAFTOVI® (encorafenib), United States Prescribing Information, 2018, Colorado, Array BioPharma Inc., a wholly owned subsidiary of Pfizer Inc., the contents of which are incorporated herein by reference in their entirety). BRAFTOVI® (encorafenib) also indicated in combination with cetuximab for the treatment of adult patients with metastatic colorectal cancer (mCRC) with a BRAF V600E mutation, after prior therapy (BRAFTOVI® (encorafenib), United States Prescribing Information, 2020, Colorado, Array BioPharma Inc., a wholly owned subsidiary of Pfizer Inc., the contents of which are incorporated herein by reference in their entirety).
Erbitux® (Cetuximab), an epidermal growth factor receptor (EGFR) antagonist, is indicated for the treatment of KRAS wild-type, EGFR-expressing, metastatic colorectal cancer in combination with FOLFIRI for first-line treatment, in combination with irinotecan in patients who are refractory to irinotecan-based chemotherapy, and as a single agent in patients who have failed oxaliplatin- and irinotecan-based chemotherapy or who are intolerant to irinotecan. Cetuximab is not indicated for treatment of Ras-mutant colorectal cancer or when the results of the Ras mutation tests are unknown.
PD-1 is recognized as an important molecule in immune regulation and the maintenance of peripheral tolerance. PD-1 is moderately expressed on naive T, B and NKT cells and up- regulated by T/B cell receptor signalling on lymphocytes, monocytes and myeloid cells (Sharpe, Arlene H et aL, The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nature Immunology (2007); 8:239-245).
Two known ligands for PD-1 , PD-L1 (B7-H1 ) and PD-L2 (B7-DC), are expressed in human cancers arising in various tissues. In large sample sets of e.g. ovarian, renal, colorectal, pancreatic, liver cancers and melanoma, it was shown that PD-L1 expression correlated with poor prognosis and reduced overall survival irrespective of subsequent treatment (Dong, Haidong et aL, Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002 Aug;8(8):793-800; Yang, Wanhua et aL, PD-1 interaction contributes to the functional suppression of T-cell responses to human uveal melanoma cells in vitro. Invest Ophthalmol Vis Sci. 2008 Jun; 49(6 (2008): 49: 2518-2525; Ghebeh, Hazem et aL, The B7-H1 (PD-L1 ) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia (2006) 8: 190-198; Hamanishi, Junzo et aL, Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl. Acad. Sci. USA (2007): 104: 3360-3365; Thompson, R Houston, and Eugene D Kwon, Significance of B7-H1 overexpression in kidney cancer. Clinical genitourin Cancer (2006): 5: 206-211 ; Nomi, Takeo et aL, Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical Cancer Research (2007);13:2151 -2157; Ohigashi, Yuichiro et aL, Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand 2 expression in human esophageal cancer. Clin. Cancer Research (2005): 11 : 2947-2953; Inman, Brant A et aL, PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer (2007): 109: 1499-1505; Shimauchi, Takatoshi et aL, Augmented expression of programmed death-1 in both neoplasmatic and nonneoplastic CD4+ T-cells in adult T-cell Leukemia/ Lymphoma. Int. J. Cancer (2007): 121 :2585-2590; Gao, Qiang et aL, Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clinical Cancer Research (2009) 15: 971-979; Nakanishi, Juro et aL, Overexpression of B7-H1 (PD-L1 ) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother. (2007) 56: 1173-1182; Hino et aL, Tumor cell expression of programmed cell death-1 is a prognostic factor for malignant melanoma. Cancer (2010): 00: 1-9). Similarly, PD-1 expression on tumor infiltrating lymphocytes was found to mark dysfunctional T cells in breast cancer and melanoma (Ghebeh, Hazem et aL, Foxp3+ tregs and B7-H1 +/PD-1 + T lymphocytes co-infiltrate the tumor tissues of high-risk breast cancer patients: implication for immunotherapy. BMC Cancer. 2008 Feb 23;8:57; Ahmadzadeh, Mojgan et aL, Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood (2009) 114: 1537-1544) and to correlate with poor prognosis in renal cancer (Thompson, R Houston et aL, PD-1 is expressed by tumor infiltrating cells and is associated with poor outcome for patients with renal carcinoma. Clinical Cancer Research (2007) 15: 1757-1761). Thus, it has been proposed that PD-L1 -expressing tumor cells interact with PD-1 -expressing T cells to attenuate T cell activation and evasion of immune surveillance, thereby contributing to an impaired immune response against the tumor.
Several monoclonal antibodies that inhibit the interaction between PD-1 and one or both of its ligands PD-L1 and PD-L2 have been approved for treating cancer. Pembrolizumab (KEYTRUDA®, Merck & Co., Inc., Rahway, NJ, USA) is a potent humanized immunoglobulin G4 (lgG4) mAb with high specificity of binding to the programmed cell death 1 (PD-1) receptor, thus inhibiting its interaction with programmed cell death ligand 1 (PD-L1) and programmed cell death ligand 2 (PD-L2). Based on preclinical in vitro data, pembrolizumab has high affinity and potent receptor blocking activity for PD-1 . Keytruda® (pembrolizumab) is indicated for the treatment of patients across a number of indications and is indicated for the first-line treatment of patients with unresectable or metastatic CRC that is microsatellite instability-high or mismatch repair deficient (MSI-H/dMMR). Pembrolizumab is the current standard of care for first line MSI-H/dMMR mCRC.
Currently, there are no first line or second line treatment options approved specifically for patients with both MSI-H/dMMR and BRAF V600E-mutant CRC. There is a necessity to develop therapies for patients with BRAF V600E-mutant CRC that is also MSI-H/dMMR that will prolong progression free survival in these subjects, increase overall survival, or both. The combination of the present invention is believed to have one or more advantages, such as improved therapeutic benefits than treatment with the therapeutic agents alone. These, and other advantages of the present invention, are apparent from the description below.
Summary of the Invention
Each of the embodiments of the present invention described below may be combined with one or more other embodiments of the present invention described herein which is not inconsistent with the embodiment(s) with which it is combined.
This invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, consisting essentially of administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with the PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with the PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging overall survival as compared to treatment with the PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging overall survival as compared to treatment with the PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging overall survival as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging overall survival as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging objective response as compared to treatment with PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging objective response as compared to treatment with PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging objective response as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging objective response as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging duration of response as compared to treatment with the PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor and an amount of a PD-1 antagonist, wherein the amounts together are effective in prolonging duration of response as compared to treatment with the PD-1 antagonist alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging duration of response as compared to treatment with pembrolizumab alone.
This invention also relates to a method of treating previously untreated BRAF V600E- mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab and an amount of pembrolizumab, wherein the amounts together are effective in prolonging duration of response as compared to treatment with pembrolizumab alone.
In one embodiment of the present invention, the encorafenib or a pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 150 mg or about 225 mg or about 300 mg calculated as the free base equivalent.
In one embodiment of the present invention, the encorafenib or a pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 300 mg calculated as the free base equivalent.
In one embodiment of the present invention, the encorafenib is a free base.
In one embodiment of the present invention, the encorafenib is formulated for oral administration. In one embodiment, the encorafenib is formulated as a capsule. In one emboidment, the capsule formulation of encorafenib comprises 50 mg of encorafenib as a free base. In one emboidment, the capsule formulation of encorafenib comprises 75 mg of encorafenib as a free base.
In one embodiment of the present invention, the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 300 mg/m2 or about 400 mg/m2 or about 500 mg/m2.
In one embodiment of the present invention, the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 500 mg/m2.
In one embodiment of the present invention, the pembrolizumab is administered as an intravenous infusion every 6 weeks at a dose of about 400 mg. In one embodiment of the present invention, the pembrolizumab is administered as an intravenous infusion every 3 weeks at a dose of about 200 mg.
In one embodiment, the pembrolizumab is administered as an intravenous infusion every three weeks at a dose of about 2 mg/kg. In one emboidment of the present invention, the colorectal cancer is metastatic or unresectable colorectal cancer. In one emboidment, the colorectal cancer is not a colorectal cancer which is RAS -mutant or for which RAS mubation status is unknown.
In one embodiment of the present invention, the subject is a human. In one embodiment, the subject is an adult patient. In one embodiment subject is a pediatric patient.
Detailed Description of the Invention
The present invention may be understood more readily by reference to the following detailed description of the preferred embodiments of the invention. It is to be understood that the terminology used herein is for the purpose of describing specific embodiments only and is not intended to be limiting. It is further to be understood that unless specifically defined herein, the terminology used herein is to be given its traditional meaning as known in the relevant art.
The term “about” when used to modify a numerically defined parameter (e.g., the dose of a encorafenib, or a pharmaceutically acceptable salt thereof, the dose of a a BRAF inhibitor and the like) means that the parameter may vary by as much as 10% above or below the stated numerical value for that parameter. For example, a dose of about 1 .0 mg once daily should be understood to mean that the dose may vary between 0.9 mg once daily and 1 .1 mg once daily.
The term “MSI-H/dMMR” refers to a cancer that is microsatellite instability-high (MSI-H), mismatch repair deficient (dMMR), or both MSI-H and dMMR.
The term “BRAF V600E-mutant, MSI-H/dMMR” refers to a cancer that has a BRAF V600E mutation and is also MSI-H, dMMR, or MSI-H and dMMR.
“Abnormal cell growth”, as used herein, unless otherwise indicated, refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). Abnormal cell growth may be benign (not cancerous), or malignant (cancerous).
The terms “cancer”, “cancerous”, and “malignant” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. As used herein “cancer” refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth. As used herein “cancer” refers to solid tumors. The term “cancer” includes, but is not limited to, a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of different type from latter one. An example of cancer for purposes of the present invention includes metastatic colorectal cancer.
The term “metastatic” (also known as “secondary cancer”) as used herein refers to a type of cancer that originates in one tissue type, but then spreads to one or more tissues outside of the (primary) cancer’s origin.
The term “patient” or “subject” refers to any single subject for which therapy is desired or that is participating in a clinical trial, epidemiological study or used as a control, including humans and mammalian veterinary patients such as cattle, horses, dogs and cats. In certain preferred embodiments, the subject is a human. A “patient” or “subject” according to the combination of this invention may have BRAF V600E mutant, MSI-H/dMMR colorectal cancer, i.e., a colorectal cancer that: 1 ) has a BRAF V600E mutation and is dMMR; 2) has a BRAF V600E mutation and is MSI-H; or 3) has a BRAF V600E mutation and is dMMR and MSI-H. A BRAF V600E mutation may be detected, for example, by analyzing a biopsy of the cancer or a peripheral blood sample (i.e., liquid biopsy) (e.g., ctDNA genetic testing) using DNA-sequencing (i.e., polymerase chain reaction (PCR)) or next generation sequencing (NGS). dMMR status may be identified, for example, by analyzing a biopsy of the cancer. MSI-H status may be identified, for example, by analyzing a biopsy of the cancer or a peripheral blood sample (i.e., liquid biopsy) (e.g., ctDNA genetic testing). In some embodiments, dMMR and/or MSI status is identified by a protein-based assay (such as by an immunoassay, such as an enzyme-linked immunosorbent assay (ELISA) or immunohistochemistry (IHC)), PCR assay (such as a real-time reverse transcriptase PCR assay) or NGS. In some embodiments, dMMR is identified by IHC. In some embodiments, MSI- H status is identified by PCR.
The term “treat” or “treating” a cancer as used herein means to administer a combination therapy according to the present invention to a subject having cancer, or diagnosed with cancer, to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastases or tumor growth, reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term "treatment", as used herein, unless otherwise indicated, refers to the act of treating as "treating" is defined immediately above. For the purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cell; inhibiting metastasis or neoplastic cells; shrinking or decreasing the size of tumor; remission of the cancer; decreasing symptoms resulting from the cancer; increasing the quality of life of the subject; delaying the progression the cancer; curing the cancer; overcoming one or more resistance mechanisms of the cancer; and / or prolonging survival of the subject. Positive therapeutic effects in cancer can be measured in a number of other ways (see, for example, W. A. Weber, J. Nucl. Med. 50:1 S-10S (200)). In some embodiments, the treatment achieved by a combination of the invention is any of the partial response (PR), complete response (CR), overall response (OR), objective response rate (ORR), progression free survival (PFS), and overall survival (OS). In some embodiments, response to a combination of the invention is any of PFS, OR or OS.
“PFS” as used herein refers to the length of time during and after the treatment of cancer, that a patient lives with the cancer but it does not get worse (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “PFS” refers the time from randomization to the date of the first documentation of progressive disease (PD) per RECIST v1 .1 or death due to any cause, whichever occurs first. In one embodiment, “PFS” refers to the length of time during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein, that a patient lives with the cancer but it does not get worse. In one embodiment, “PFS” refers to the length of time during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein, that a patient lives with the cancer but it does not get worse as compared to treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer with pembrolizumab alone.
“OS” as used herein refers to a prolongation in life expectancy as compared to untreated subjects (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “OS” refers to the time from the date of randomization to the date of death due to any cause. In one embodiment, “OS” refers to the prolongation of life expectancy during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein. In one embodiment, “OS” refers to the prolongation of life expectancy during and after the treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone. “OR” as used herein refers to confirmed complete response (CR) or confirmed PR (partial response), based on investigator assessment per RECIST v1 .1 , from the date of randomization until the date of the first documentation of progressive disease (PD), death or start of new anticancer therapy. In one embodiment, “OR” refers to confirmed complete response (CR) or confirmed PR (partial response) during and after the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone.
“ORR” as used herein is defined as the percentage of people in a study or treatment group who have a partial or complete response to the treatment within a certain period of time (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “ORR” refers to the percentage of people in a study or treatment group who have a partial or complete response to the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone.
“CR” as used herein refers to the disappearance of all signs of cancer (e.g., tumor lesions) in the body in response to treatment (also referred to as complete remission) (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “CR” refers to the disappearance of all signs of cancer (e.g., tumor lesions) in the body in response to the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer with pembrolizumab alone.
“PR” as used herein refers to a decrease in the size of a tumor as determined per RECIST v1 .1 , or in the extent of cancer in the body, in response to treatment (also referred to as partial remission) (National Cancer Institute, Dictionary of Cancer Terms). In one embodiment, “PR” refers to a decrease in the size of a tumor as determined per RECIST v1 .1 , or in the extent of cancer in the body, in response to treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer according to any of the methods described herein as compared to treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer with pembrolizumab alone. “PD” as used herein refers to a cancer that is growing, spreading, or getting worse (National Cancer Institute, Dictionary of Cancer Terms).
Response to a combination of the invention, may be assessed using Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1 ) response criteria. The treatment regimen for a combination of the invention that is effective to treat a subject in need thereof may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the subject. While an embodiment of any of the aspects of the invention may not be effective in achieving a positive therapeutic effect in every subject, it should do so in a statistically significant number of subjects as determined by any statistical test known in the art such as, but not limited to, the Cox log-rank test, the Cochran- Mantel-Haenszel log-rank test, the Student’s t-test, the chi2-test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstrat-test and the Wilcon on-test.
The terms “treatment regimen” and “dosing regimen” are used interchangeably to refer to the dose and timing of administration of each therapeutic agent in a combination of the invention.
“Ameliorating” means a lessening or improvement of one or more symptoms as compared to not administering a therapeutic agent of a method or regimen of the invention. “Ameliorating” also includes shortening or reduction in duration of a symptom.
An “effective dosage,” “effective amount,” "therapeutically effective amount," or “therapeutically effective dosage” of a drug, therapeutic agent, or pharmaceutical composition is an amount to affect any one or more beneficial or desired, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use in reference to the treatment of cancer, an effective amount refers to that amount which has the effect of (1 ) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, (4) relieving to some extent (or, preferably, eliminating) one or more signs or symptoms associated with the cancer, (5) enhancing the effect of another medication, and / or (6) delaying the progression of the disease of the subject. In one embodiment, an effective amount refers to that amount which has the effect of prolonging one or more of progression-free survival, overall survival, objective response, and/or duration of response. An effective dosage can be administered in one or more administrations. As is understood in the clinical context, dosage of a therapeutically effective amount of a compound or pharmaceutical composition thereof may be achieved in conjunction with another therapy. Thus, a therapeutically effective amount may be considered in the context of administering one or more therapies (e.g., one or more anticancer agents), and a single agent may be considered to be given in a therapeutically effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
“Tumor” as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size, and includes primary tumors and secondary neoplasms. A solid tumor is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Examples of solid tumors are sarcomas, carcinomas, and lymphomas. Leukaemia’s (cancers of the blood) generally do not form solid tumors (National Cancer Institute, Dictionary of Cancer Terms).
The term “tumor size” refers to the total size of the tumor which can be measured as the length and width of a tumor. Tumor size may be determined by a variety of methods known in the art, such as, e.g., by measuring the dimensions of tumor(s) upon removal from the subject, e.g., using callipers, or while in the body using imaging techniques, e.g., ultrasound, a computed tomography (CT) scan (also referred to as a CAT scan), or magnetic resonance imaging (MRI).
As used herein, the term “previously untreated” means the subject has not received prior systemic regimens for metastatic CRC. In one embodiment, “previously untreated” does not include subjects with early-stage cancer (e.g., Stages l-lll) treated with surgery followed by chemotherapy (e.g., treatment in the adjuvant setting) or who have received prior systemic neoadjuvant therapy with or without radiation who present with new lesions or evidence of recurrence of the cancer during or within 6 months of the last dose of chemotherapy.
The term “systemic therapy” as used herein means treatment using substances that travel through the bloodstream, reaching and affecting cells all over the body (National Cancer Institute, Dictionary of Cancer Terms). Examples of systemic regimens (i.e., therapies) include BRAF inhibitors (e.g., encorafenib, dabrafenib, vemurafenib, or XL281/BMS-908662), EGFR inhibitors (e.g., cetuximab or panitumumab), and immune checkpoint inhibitors such as PD-1 antagonists, or agents directed to another stimulatory or co-inhibitory T-cell receptor (e.g., CTLA-4, 0X40, or CD137). The term “neoadjuvant” as used herein means a treatment given as a first step to shrink a tumor before the main treatment (e.g., a combination therapy or the present invention). Examples of neoadjuvant treatment include chemotherapy, radiation therapy and hormone therapy (National Cancer Institute, Dictionary of Cancer Terms).
As used herein, the term “antibody” refers to any form of antibody that exhibits the desired biological or binding activity. Thus, it is used in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized, fully human antibodies, chimeric antibodies and camelized single domain antibodies. “Parental antibodies” are antibodies obtained by exposure of an immune system to an antigen prior to modification of the antibodies for an intended use, such as humanization of an antibody for use as a human therapeutic.
In general, the basic antibody structural unit comprises a tetramer. Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function. Typically, human light chains are classified as kappa and lambda light chains. Furthermore, human heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids. See generally, Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989).
The variable regions of each light/heavy chain pair form the antibody binding site. Thus, in general, an intact antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are, in general, the same.
Typically, the variable domains of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope. In general, from N-terminal to C- terminal, both light and heavy chains variable domains comprise FR1 , CDR1 , FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al.; National Institutes of Health, Bethesda, Md. ; 5th ed.; NIH Publ. No. 91 -3242 (1991 ); Kabat (1978) Adv. Prot. Chem. 32:1 -75; Kabat, et al., (1977) J. Biol. Chem. 252:6609-6616; Chothia, et al., (1987) J Mol. Biol. 196:901 -917 or Chothia, et al., (1989) Nature 342:878-883.
As used herein, unless otherwise indicated, “antibody fragment” or “antigen binding fragment” refers to antigen binding fragments of an antibody, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full-length antibody, e.g. fragments that retain one or more CDR regions, e.g. all six CDRs. Examples of antibody binding fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and multispecific antibodies formed from antibody fragments.
An antibody that “specifically binds to” a specified target protein is an antibody that exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity. An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g. without producing undesired results such as false positives. Antibodies, or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20- times greater, and most preferably at least 100-times greater than the affinity with non-target proteins. As used herein, an antibody is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human PD-1 or human PD-L1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
“CDR” or “CDRs” as used herein means complementarity determining region(s) in a immunoglobulin variable region, defined using the Kabat numbering system, unless otherwise indicated.
“Conservatively modified variants” or “conservative substitution” refers to substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g. charge, sidechain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity or other desired property of the protein, such as antigen affinity and/or specificity. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity. Exemplary conservative substitutions are set forth in Table 1 below.
TABLE 1 . Exemplary Conservative Amino Acid Substitutions
Figure imgf000022_0001
“Framework region” or “FR” as used herein means the immunoglobulin variable regions excluding the CDR regions.
“Kabat” as used herein means an immunoglobulin alignment and numbering system pioneered by Elvin A. Kabat ((1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.).
“Monoclonal antibody” or “mAb” or “Mab”, as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731 .
“PD-1 antagonist” means any chemical compound or biological molecule that blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or Natural Killer T cell) and in specific embodiments also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1 . Alternative names or synonyms for PD-1 and its ligands include: PDCD1 , PD1 , CD279 and SLEB2 for PD-1 ; PDCD1 L1 , PDL1 , B7H1 , B7-4, CD274 and B7-H for PD-L1 ; and PDCD1 L2, PDL2, B7-DC, Btdc and CD273 for PD-L2. In any of the treatment method, medicaments and uses of the present invention in which a human individual is being treated, the PD-1 antagonist blocks binding of human PD-L1 to human PD-1 , and in specific embodiments blocks binding of both human PD-L1 and PD-L2 to human PD-1. Human PD-1 amino acid sequences can be found in NCBI Locus No.: NP 005009. Human PD- L1 and PD-L2 amino acid sequences can be found in NCBI Locus No.: NP 054862 and NP 079515, respectively.
“Pembrolizumab” (formerly known as MK-3475, SCH 900475 and lambrolizumab) alternatively referred to herein as “pembro,” is a humanized lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 2, pages 161-162 (2013) and which comprises the heavy and light chain amino acid sequences and CDRs described in Table 2. Pembrolizumab has been approved by the U.S. FDA as described in the Prescribing Information for KEYTRUDA™ (Merck & Co., Inc., Rahway, NJ, USA; initial U.S. approval 2014, updated March 2021).
As used herein, a “pembrolizumab variant” or “a variant thereof” pertaining to a pembrolizumab sequence means a monoclonal antibody that comprises heavy chain and light chain sequences that are substantially identical to those in pembrolizumab, except for having three, two or one conservative amino acid substitutions at positions that are located outside of the light chain CDRs and six, five, four, three, two or one conservative amino acid substitutions that are located outside of the heavy chain CDRs, e.g., the variant positions are located in the FR regions or the constant region, and optionally has a deletion of the C-terminal lysine residue of the heavy chain. In other words, pembrolizumab and a pembrolizumab variant comprise identical CDR sequences, but differ from each other due to having a conservative amino acid substitution at no more than three or six other positions in their full-length light and heavy chain sequences, respectively. A pembrolizumab variant is substantially the same as pembrolizumab with respect to the following properties: binding affinity to PD-1 and ability to block the binding of each of PD-L1 and PD-L2 to PD-1.
Therapeutic Methods and Uses
The methods and combination therapies of the present invention are useful for treating cancer. In one embodiment, the cancer is a previously untreated BRAF V600E mutant, MSI- H/dMMR colorectal cancer. Colorectal cancer is also referred to as colorectal adenocarcinoma.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of a BRAF inhibitor, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, the BRAF inhibitor is selected from encorafenib, dabrafenib, vemurafenib, N-[3-(5-chloro-1 H-pyrrolo[2,3-b]pyridin-3-ylcarbonyl)-2,4-difluorophenyl]propane-
1 -sulfonamide (PLX4720), and (3R)-N-(3-[[5-(2-cyclopropylpyrimidin-5-yl)-1 H-pyrrolo[2,3- b]pyridin-3-yl]carbonyl]-2 ,4-dif luorophenyl)-3-f luoropyrrolidine- 1 -sulfonamide (PLX8394), and pharmaceutically acceptable salts thereof, compounds disclosed in International Application No. PCT/IB2020/055992, published December 30, 2020 as PCT Publication No. WO 2020/261 156 A1 , including, for example, a compound selected from N-(3-((3,5-dimethyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)-2,4-difluorophenyl)propane-1 -sulfonamide, N-(2-chloro-3-((3,5- dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-3-fluoropropane-1 -sulfonamide, N-(2- chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4,5-difluorophenyl)propane-1 - sulfonamide, N-(2-chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4- fluorophenyl)propane-1 -sulfonamide, N-(2-chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin- 6-yl)amino)-4-fluorophenyl)-3-fluoropropane-1 -sulfonamide, N-(2-chloro-4-fluoro-3-((5-methyl-3- (methyl-d3)-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-phenyl)-3-fluoropropane-1 -sulfonamide, N- {2-chloro-3-[(3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)oxy]-4-fluorophenyl}propane-1 - sulfonamide, N-(3-chloro-4-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)oxy)-5-fluoropyridin-
2-yl)propane-1 -sulfonamide; and N-{2-chloro-3-[(3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6- yl)oxy]-4-fluorophenyl}-3-fluoropropane-1 -sulfonamide, and pharmaceutically acceptable salt thereof; and compounds disclosed in PCT Publication No. WO 2021/250521 , published December 16, 2021 , including, for example, N-(2-chloro-4-fluoro-3-((5-fluoro-3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-2-azabicyclo[2.1 ,1]hexane-2-sulfonamide, (R)-N-(2- chloro-4-fluoro-3-((5-fluoro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-3- fluoropyrrolidine-1 -sulfonamide, and N-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3-fluoroazetidine-1 -sulfonamide, and pharmaceutically acceptable salt thereof.
In one embodiment, the BRAF inhibitor is selected from encorafenib or a pharmaceutically acceptable salt thereof, /V-(2-chloro-3-((3,5-dimethyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4- fluorophenyl)-3-fluoropropane-1 -sulfonamide or a pharmaceutically acceptable salt thereof, and N-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3- fluoroazetidine-1 -sulfonamide or a pharmaceutically acceptable salt thereof. In one embodiment, the BRAF inhibitor is encorafenib or a pharmaceutically acceptable salt thereof. In one embodiment, the BRAF inhibitor is encorafenib as a free base.
In one embodiment, the EGFR inhibitor is selected from cetuximab (Erbitux®), panitumumab (Vectibix®), osimertinib (Tagrisso®), erlotinib (Tarceva®), gefitinib (Iressa®), necitumumab (Portrazza™), neratinib (Nerlynx®), lapatinib (Tykerb®), vandetanib (Caprelsa®), brigatinib (Alunbrig®). In one embodiment, the EGFR inhibitor is cetuximab.
In one embodiment, the PD-1 antagonist useful in the treatment, medicaments and uses of the present invention include a monoclonal antibody (mAb), or antigen binding fragment thereof, that specifically binds to PD-1 or PD-L1 , and preferably specifically binds to human PD- 1 or human PD-L1 . The mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region. In some embodiments the human constant region is selected from the group consisting of IgG 1 , lgG2, lgG3 and lgG4 constant regions, and in some embodiments, the human constant region is an IgG 1 or lgG4 constant region. In some embodiments, the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab')2, scFv and Fv fragments.
Examples of mAbs that bind to human PD-1 , and useful in the treatment method, medicaments and uses of the present invention, are described in U.S. patent Nos. US 7488802, US 7521051 , US 8008449, US 8354509, and US 8168757, and International application publn. Nos. WO 2004/004771 , WO 2004/072286, WO 2004/056875, and WO 2008/156712, and US. Application publication No. US 2011/0271358. Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include: pembrolizumab (also known as MK-3475), a humanized lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 2, pages 161-162 (2013) and that comprises the heavy and light chain amino acid sequences shown in Table 2; nivolumab (BMS-936558), a human lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 1 , pages 68-69 (2013) and that comprises the heavy and light chain amino acid sequences shown in Table 2; the humanized antibodies h409A11 , h409A16 and h409A17, which are described in WO2008/156712, and AMP-514, which is being developed by Medlmmune; cemiplimab; camrelizumab; sintilimab; tislelizumab; and toripalimab. Additional anti-PD-1 antibodies contemplated for use herein include MEDI0680 (U.S. Patent no. 8609089), BGB-A317 (U.S. Patent publ. no. 2015/0079109), INCSHR1210 (SHR-1210) (PCT International application publ. no. WO2015/085847), REGN-2810 (PCT International application publ. no. WO2015/112800), PDR001 (PCT International application publ. no. WO2015/112900), TSR-042 (ANB011 ) (PCT International application publ. no. WO2014/179664) and STI-1110 (PCT International application publ. no. WO2014/194302).
Examples of mAbs that bind to human PD-L1 , and useful in the treatment method, medicaments and uses of the present invention, are described in US8383796. Specific antihuman PD-L1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include BMS-936559, MEDI4736, and MSB0010718C.
In some embodiments, the PD-1 antagonist is pembrolizumab (KEYTRUDA™, Merck & Co., Inc., Rahway, NJ, USA), nivolumab (OPDIVO™, Bristol-Myers Squibb Company, Princeton, NJ, USA), atezolizumab (TECENTRIQ™, Genentech, San Francisco, CA, USA), durvalumab (IMFINZI™, AstraZeneca Pharmaceuticals LP, Wilmington, DE), cemiplimab (LIBTAYO™, Regeneron Pharmaceuticals, Tarrytown, NY, USA) avelumab (BAVENCIO™, Merck KGaA, Darmstadt, Germany) or dostarlimab (JEMPERLI™, GlaxoSmithKline LLC, Philadelphia, PA). In other embodiments, the PD-1 antagonist is pidilizumab (U.S. Pat. No. 7,332,582), AMP-514 (Medlmmune LLC, Gaithersburg, MD, USA), PDR001 (U.S. Pat. No. 9,683,048), BGB-A317 (U.S. Pat. No. 8,735,553), or MGA012 (MacroGenics, Rockville, MD).
In one embodiment, the PD-1 antagonist useful in the methods of the invention is an anti-PD-1 antibody that blocks the binding of PD-1 to PD-L1 and PD-L2. In some embodiments of the treatment methods, medicaments and uses of the present invention, the PD-1 antagonist is a monoclonal antibody, or antigen binding fragment thereof, that comprises: (a) a light chain variable region comprising light chain CDR1 , CDR2 and CDR3 of SEQ ID NOs: 1 , 2 and 3, respectively and (b) a heavy chain variable region comprising heavy chain CDR1 , CDR2 and CDR3 of SEQ ID NOs: 6, 7 and 8, respectively.
In other embodiments of the treatment methods, medicaments and uses of the present invention, the PD-1 antagonist is a monoclonal antibody, or antigen binding fragment thereof, that specifically binds to human PD-1 and comprises (a) a heavy chain variable region comprising SEQ ID NO:9 or a variant thereof, and (b) a light chain variable region comprising SEQ ID NO:4 or a variant thereof. A variant of a heavy chain variable region sequence is identical to the reference sequence except having up to six conservative amino acid substitutions in the framework region (i.e. , outside of the CDRs). A variant of a light chain variable region sequence is identical to the reference sequence except having up to three conservative amino acid substitutions in the framework region (i.e., outside of the CDRs).
In another embodiment of the treatment methods, medicaments and uses of the present invention, the PD-1 antagonist is a monoclonal antibody that specifically binds to human PD-1 and comprises (a) a heavy chain comprising SEQ ID NO: 10 and (b) a light chain comprising SEQ ID NO:5. In one embodiment, the PD-1 antagonist is an anti-PD-1 antibody that comprises two heavy chains and two light chains, and wherein the heavy and light chains comprise the amino acid sequences in SEQ ID NQ:10 and SEQ ID NO:5, respectively.
In all of the above treatment methods, medicaments and uses, the PD-1 antagonist inhibits the binding of PD-L1 to PD-1 , and in specific embodiments also inhibits the binding of PD-L2 to PD-1 . In some embodiments of the above treatment methods, medicaments and uses, the PD-1 antagonist is a monoclonal antibody, or an antigen binding fragment thereof, that specifically binds to PD-1 or to PD-L1 and blocks the binding of PD-L1 to PD-1 .
Table 2 below provides a list of the amino acid sequences of exemplary anti-PD-1 mAbs for use in the treatment method, medicaments and uses of the present invention. Table 2. Exemplary PD-1 Antibody Sequences
Figure imgf000029_0001
Table 3. Additional PD-1 Antibodies and Antigen Binding Fragments Useful in the Formulations, Methods and Uses of the Invention.
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
In one embodiment, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain constant region, e.g. a human constant region, such as g1 , g2, g3, or g4 human heavy chain constant region or a variant thereof. In another embodiment, the anti- PD-1 antibody or antigen-binding fragment thereof comprises a light chain constant region, e.g. a human light chain constant region, such as lambda or kappa human light chain region or a variant thereof. By way of example, and not limitation, the human heavy chain constant region can be g4 and the human light chain constant region can be kappa. In an alternative embodiment, the Fc region of the antibody is g4 with a Ser228Pro mutation (Schuurman, J et. al., Mol. Immunol. 38: 1 -8, 2001 ). In some embodiments, different constant domains may be appended to humanized VL and VH regions derived from the CDRs provided herein. For example, if a particular intended use of an antibody (or fragment) of the present invention were to call for altered effector functions, a heavy chain constant domain other than human IgG 1 may be used, or hybrid lgG1/lgG4 may be utilized. Although human lgG1 antibodies provide for long half-life and for effector functions, such as complement activation and antibody-dependent cellular cytotoxicity, such activities may not be desirable for all uses of the antibody. In such instances a human lgG4 constant domain, for example, may be used. The present invention includes the use of anti-PD-1 antibodies or antigen-binding fragments thereof which comprise an lgG4 constant domain. In one embodiment, the lgG4 constant domain can differ from the native human lgG4 constant domain (Swiss-Prot Accession No. P01861 .1) at a position corresponding to position 228 in the Ell system and position 241 in the KABAT system, where the native Ser108 is replaced with Pro, in order to prevent a potential inter-chain disulfide bond between Cys106 and Cys109 (corresponding to positions Cys 226 and Cys 229 in the EU system and positions Cys 239 and Cys 242 in the KABAT system) that could interfere with proper intra-chain disulfide bond formation. See Angal et al. (1993) Mol. ImunoL 30:105. In other instances, a modified IgG 1 constant domain which has been modified to increase half-life or reduce effector function can be used.
In another embodiment, the PD-1 antagonist is an antibody or antigen binding protein that has a variable light domain and/or a variable heavy domain with at least 95%, 90%, 85%, 80%, 75% or 50% sequence identity to one of the variable light domains or variable heavy domains described above, and exhibits specific binding to PD-1 . In another embodiment of the methods of treatment of the invention, the PD-1 antagonist is an antibody or antigen binding protein comprising variable light and variable heavy domains having up to 1 , 2, 3, 4, or 5 or more amino acid substitutions, and exhibits specific binding to PD-1
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of a PD-1 antagonist, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of an EGFR inhibitor, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying one or both of dMMR status from a biopsy of the cancer from the subject and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
In one embodiment, this invention relates to a BRAF inhibitor for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the BRAF inhibitor is used in combination with an EGFR inhibitor and a PD-1 antagonist.
In one embodiment, this invention relates to an EGFR inhibitor for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the EGFR is used in combination with a BRAF inhibitor and a PD-1 antagonist.
In another aspect, this invention relates to a PD-1 antagonist for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the PD-1 antagonist is used in combination with a BRAF inhibitor and an EGFR inhibitor.
In another aspect, this invention relates to encorafenib, or a pharmaceutically acceptable salt thereof, for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the encorafenib, or a pharmaceutically acceptable salt thereof, is used in combination with cetuximab and pembrolizumab.
In another aspect, this invention relates to cetuximab for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the cetuximab is used in combination with encorafenib, or a pharmaceutically acceptable salt thereof, and pembrolizumab.
In another aspect, this invention relates to pembrolizumab for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the pembrolizumab is used in combination with encorafenib, or a pharmaceutically acceptable salt thereof, and cetuximab.
In another aspect, this invention relates to the use of encorafenib, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the medicament used in combination with cetuximab and pembrolizumab.
In another aspect, this invention relates to the use of cetuximab in the manufacture of a medicament for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the medicament used in combination with encorafenib or a pharmaceutically acceptable salt thereof, and pembrolizumab.
In another aspect, this invention relates to the use of pembrolizumab in the manufacture of a medicament for the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the medicament used in combination with encorafenib or a pharmaceutically acceptable salt thereof, and cetuximab.
In another aspect, this invention relates to a pharmaceutical composition comprising encorafenib, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient, for use in the treatment of previously untreated BRAF V600E-mutant, MSI- H/dMMR colorectal cancer in a subject in need thereof, wherein the pharmaceutical composition comprising the encorafenib, or a pharmaceutically acceptable salt thereof, is used in combination with a pharmaceutical composition comprising cetuximab and at least one pharmaceutically acceptable excipient, and a pharmaceutical composition comprising pembrolizumab and at least one pharmaceutically acceptable excipient.
In another aspect, this invention relates to a pharmaceutical composition comprising cetuximab and at least one pharmaceutically acceptable excipient, for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the pharmaceutical composition comprising the cetuximab is used in combination with a pharmaceutical composition comprising encorafenib or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient, and a pharmaceutical composition comprising pembrolizumab and at least one pharmaceutically acceptable excipient.
In another aspect, this invention relates to a pharmaceutical composition comprising pembrolizumab and at least one pharmaceutically acceptable excipient for use in the treatment of previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, wherein the pharmaceutical composition comprising the pembrolizumab is used in combination with a pharmaceutical composition comprising encorafenib or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient, and a pharmaceutical composition comprising cetuximab and at least one pharmaceutically acceptable excipient.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, the colorectal cancer is BRAF V600E-mutant, MSI-H colorectal cancer.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, the colorectal cancer is BRAF V600E-mutant, dMMR colorectal cancer.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, the colorectal cancer is BRAF V600E-mutant, MSI-H, dMMR colorectal cancer.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, the colorectal cancer is metastatic or unresectable colorectal cancer. In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, progression-free survival is prolonged as compared to treatment with pembrolizumab alone.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, overall survival is prolonged as compared to treatment with pembrolizumab alone.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, objective response is prolonged as compared to treatment with pembrolizumab alone.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, duration of response is prolonged as compared to treatment with pembrolizumab alone.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, the encorafenib is the free base.
In one embodiment of any of the methods, uses or pharmaceutical compositions of the invention, the subject is a mammal.
In one embodiment of any of the methods or pharmaceutical compositions of the invention, the subject is a human.
In one embodiment of any of the methods of the invention, the BRAF V600E mutation is detected in tumor tissue by PCR or NGS.
In one embodiment of any of the methods of the invention, dMMR status is identified by IHC.
In one embodiment of any of the methods of the invention, MSI-H status is identified by DNA-based testing (PCR).
Dosaae Forms and Reaimens
Each therapeutic agent of the methods and combination therapies of the present invention may be administered either alone, or in a medicament (also referred to herein as a pharmaceutical composition) comprising the therapeutic agent and at least one pharmaceutically acceptable excipient, according to pharmaceutical practice.
As used herein, the term “combination therapy” refers to the administration of each therapeutic agent of the combination therapy of the invention, either alone or in a medicament, either sequentially or concurrently, wherein the agents are dosed in amounts which together are effective in treating the cancer.
As used herein, the term “sequential” or “sequentially” refers to the administration of each therapeutic agent of the combination therapy of the invention, either alone or in a medicament, one after the other, wherein each therapeutic agent can be administered in any order and with variable intervening time limits. Sequential administration is particularly useful when the therapeutic agents in the combination therapy are in different dosage forms, for example, one agent is a tablet and the second agent and/or third agent is a sterile liquid, and/or the therapeutic agents are administered by different routes of administration, for example one agent is administered orally and the second agent and/or third agent is administered intravenously, and/or the therapeutic agents are administered according to different dosing schedules, for example, one agent is administered daily, and the second agent and/or third agent is administered less frequently such as weekly, every two weeks, or every 6 weeks.
As used herein, the term “concurrently” refers to the administration of each therapeutic agent in the combination therapy of the invention, either alone or in separate medicaments, wherein the second and third therapeutic agents are administered immediately after the first therapeutic agent, but that the therapeutic agents can be administered in any order.
In one embodiment of the present invention, the encorafenib is formulated for oral administration. In one embodiment, the encorafenib is formulated as a capsule. In one emboidment, the capsule formulation of encorafenib comprises 50 mg of encorafenib as a free base. In one emboidment, the capsule formulation of encorafenib comprises 75 mg of encorafenib as a free base.
In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 150 mg or about 225 mg or about 300 mg calculated as the free base equivalent. In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent. In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent. In one embodiment, an effective dosage of encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent.
In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m2 or about 400 mg/m2 or about 500 mg/m2. In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m2. In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m2. In one embodiment, an effective dosage of cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m2. In one embodiment, cetuximab is administered as a 120-minute intravenous infusion. In one embodiment, the subject is administered a premedication (e.g., antihistamine or antipyretic) prior to administration of cetuximab.
In one embodiment, an effective dosage of pembrolizumab is administered at a dose of about 400 mg every 6 weeks (Q6W). In one embodiment, an effective dosage of pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W). In one embodiment, pembrolizumab is administered as a 30-minute (-5 minutes /+10 minutes) intravenous infusion. In one embodiment, the selected dose of pembrolizumab is administered by IV infusion over a time period of between 25 and 40 minutes, or about 30 minutes. In one embodiment, the subject is administered a premedication (e.g., antihistamine or antipyretic) prior to administration of pembrolizumab. In one embodiment, the selected dose of pembrolizumab is administered by subcutaneous injection.
In one embodiment, an effective dosage of pembrolizumab is administered as an intravenous infusion at a dose of about 200 mg every 3 weeks (Q3W). In one embodiment, the method comprises administering 2 mg/kg of pembrolizumab to the patient about every three weeks. In particular embodiments, the patient is a pediatric patient.
In some embodiments, at least one of the therapeutic agents in the combination therapy is administered using the same dosage regimen (dose, frequency, and duration of treatment) that is typically employed when the agent is used as a monotherapy for treating the same cancer. In other embodiments, the subject received a lower total amount of at least one of the therapeutic agents in the combination therapy than when the same agent is used as a monotherapy, for example a lower dose of therapeutic agent, a reduced frequency of dosing and / or a shorter duration of dosing.
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 150 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W). In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 225 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 300 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 400 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment of the present invention, encorafenib, or a pharmaceutically acceptable salt thereof, is administered orally as a once daily (QD) dosage of about 300 mg calculated as the free base equivalent, cetuximab is administered as an intravenous infusion every two weeks (Q2W) at a dose of about 500 mg/m2, and pembrolizumab is administered as an intravenous infusion at a dose of about 400 mg every 6 weeks (Q6W).
In one embodiment, when all three of encorafenib or a pharmaceutically acceptable salt thereof, cetuximab and pembrolizumab are administered on the same day, the encorafenib or a pharmaceutically acceptable salt thereof is administered prior to administration of the pembrolizumab, which is then followed by administration of the cetuximab.
In one embodiment, when the cetuximab and the pembrolizumab are administered on the same day, the cetuximab is administered about 30-60 minutes after administration of the pembrolizumab. Dosage regimens may be adjusted to provide the optimum desired response. For example, a therapeutic agent of the combination therapy of the present invention may be administered as a single bolus, as several divided doses administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be particularly advantageous to formulate a therapeutic agent in a dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical excipient. The specification for the dosage unit forms of the invention may be dictated by and directly dependent on (a) the unique characteristics of the therapeutic agent and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
Thus, the skilled artisan would appreciate, based upon the disclosure provided herein, that the dose and dosing regimen is adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose may be readily established, and the effective amount providing a detectable therapeutic benefit to a subject may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the subject. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a subject in practicing the present invention.
It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, taking into consideration factors such as the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. The dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values. Thus, the present invention encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regimens for administration of the therapeutic agent are well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
Repetition of the administration or dosing regimens, or adjustment of the administration or dosing regimen may be conducted as necessary to achieve the desired treatment. A “continuous dosing schedule” as used herein is an administration or dosing regimen without dose interruptions, e.g., without days off treatment. Repetition of 21 - or 28-day treatment cycles without dose interruptions between the treatment cycles is an example of a continuous dosing schedule. In an embodiment, the compounds of the combination of the present invention can be administered in a continuous dosing schedule.
Pharmaceutical Compositions and Routes of Administration
A "pharmaceutical composition" refers to a mixture of one or more of the compounds of the invention, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof as an active ingredient, and at least one pharmaceutically acceptable excipient.
The term “excipient” is used herein to describe any ingredient other than the therapeutic agents of the combination therapy. The choice of excipient will to a large extent depend on factors such as the mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
As used herein, "excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, carriers, diluents and the like that are physiologically compatible. Examples of excipients include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof, and may include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol, or sorbitol in the composition. Examples of excipients also include various organic solvents (such as hydrates and solvates). The pharmaceutical compositions may, if desired, contain additional excipients such as flavorings, binders/binding agents, lubricating agents, disintegrants, sweetening or flavoring agents, coloring matters or dyes, and the like. For example, for oral administration, tablets containing various excipients, such as citric acid may be employed together with various disintegrants such as starch, alginic acid and certain complex silicates and with binding agents such as sucrose, gelatin and acacia. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes. Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules. Non-limiting examples of excipients, therefore, also include lactose or milk sugar and high molecular weight polyethylene glycols. When aqueous suspensions or elixirs are desired for oral administration of the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with additional excipients such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
Examples of excipients also include pharmaceutically acceptable substances such as wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives, or buffers, which enhance the shelf life or effectiveness of the compound.
The compositions of each of the therapeutic agents of the combination of the invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, capsules, pills, powders, liposomes and suppositories. The form depends on the intended mode of administration and therapeutic application.
Administration of a composition comprising a therapeutic agent used in the combination of the invention may be oral, parenteral, topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), or pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal),
Oral administration of a solid dosage form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount the therapeutic agent. In another embodiment, the oral administration may be in a powder or granule form. In another embodiment, the oral dosage form is sub-lingual, such as, for example, a lozenge. In such solid dosage forms, the therapeutic agent is ordinarily combined with one or more adjuvants. Such capsules or tablets may comprise a controlled release formulation. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agents or may be prepared with enteric coatings. In another embodiment, oral administration may be in a liquid dosage form. Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as one or more of wetting, emulsifying, suspending, flavoring (e.g., sweetening), or perfuming agents.
In another embodiment, the therapeutic agent may be administered in a parenteral dosage form. "Parenteral administration" includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion. Injectable preparations (i.e. , sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using one or more of suitable dispersing, wetting agents, or suspending agents.
In another embodiment, the therapeutic agent may be administered in a topical dosage form. "Topical administration" includes, for example, dermal and transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. A topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of this invention are administered by a transdermal device, administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages and microemulsions. Liposomes may also be used. Typical excipients include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, B. C. Finnin and T. M. Morgan, J. Pharm. Sci., vol. 88, pp. 955-958, 1999.
Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in a suitable excipient. A typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methylcellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
For intranasal administration, the therapeutic agent may be conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant. Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1 ,1 ,1 ,2- tetrafluoroethane or 1 ,1 ,1 ,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
In another embodiment, the therapeutic agent may be administered as a rectal dosage form. Such rectal dosage form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
Other excipients and modes of administration known in the pharmaceutical art may also be used. Pharmaceutical compositions of the therapeutic agents may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures. The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks. Formulation of drugs is discussed in, for example, Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman et aL, Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe et aL, Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999.
Acceptable excipients are nontoxic to subjects at the dosages and concentrations employed, and may comprise one or more of the following: 1 ) buffers such as phosphate, citrate, or other organic acids; 2) salts such as sodium chloride; 3) antioxidants such as ascorbic acid or methionine; 4) preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol; 5) alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, or m-cresol; 6) low molecular weight (less than about 10 residues) polypeptides; 7) proteins such as serum albumin, gelatin, or immunoglobulins; 8) hydrophilic polymers such as polyvinylpyrrolidone; 9) amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; 10) monosaccharides, disaccharides, or other carbohydrates including glucose, mannose, or dextrins; 1 1 ) chelating agents such as EDTA; 12) sugars such as sucrose, mannitol, trehalose or sorbitol; 13) salt-forming counter-ions such as sodium, metal complexes (e.g., Zn- protein complexes), or 14) non-ionic surfactants such as polysorbates (e.g., polysorbate 20 or polysorbate 80), poloxamers or polyethylene glycol (PEG).
Liposome containing a therapeutic agent may be prepared by methods known in the art (See, for example, Chang, H.L; Yeh, M.K.; Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy; Int J Nanomedicine 2012; 7; 49-60). Particularly useful liposomes may be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
A therapeutic agent may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington, The Science and Practice of Pharmacy, 20th Ed., Mack Publishing (2000).
Sustained-release preparations may be used. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing a therapeutic agent, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate) or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as those used in leuprolide acetate for depot suspension (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3-hydroxybutyric acid.
Suitable emulsions may be prepared using commercially available fat emulsions, such as a lipid emulsions comprising soybean oil, a fat emulsion for intravenous administration (e.g., comprising safflower oil, soybean oil, egg phosphatides and glycerin in water), emulsions containing soya bean oil and medium-chain triglycerides, and lipid emulsions of cottonseed oil. The active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g., egg phospholipids, soybean phospholipids or soybean lecithin) and water. It will be appreciated that other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion. Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%. The fat emulsion may comprise fat droplets between 0.1 and 1.0 pm, particularly 0.1 and 0.5 pm, and have a pH in the range of 5.5 to 8.0.
For example, the emulsion compositions may be those prepared by mixing a therapeutic agent with a lipid emulsion comprising soybean oil or the components thereof (soybean oil, egg phospholipids, glycerol and water).
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulized by use of gases. Nebulized solutions may be breathed directly from the nebulizing device, or the nebulizing device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
A drug product intermediate (DPI) is a partly processed material that must undergo further processing steps before it becomes bulk drug product. A therapeutic agent of the invention may be formulated into drug product intermediate DPI containing the active ingredient in a higher free energy form than the crystalline form. One reason to use a DPI is to improve oral absorption characteristics due to low solubility, slow dissolution, improved mass transport through the mucus layer adjacent to the epithelial cells, and in some cases, limitations due to biological barriers such as metabolism and transporters. Other reasons may include improved solid-state stability and downstream manufacturability. In one embodiment, the drug product intermediate contains a therapeutic agent isolated and stabilized in the amorphous state (for example, amorphous solid dispersions (ASDs)). There are many techniques known in the art to manufacture ASD’s that produce material suitable for integration into a bulk drug product, for example, spray dried dispersions (SDD’s), melt extrudates (often referred to as HME’s), co-precipitates, amorphous drug nanoparticles, and nano-adsorbates. In one embodiment amorphous solid dispersions comprise a therapeutic agent and a polymer excipient. Other excipients as well as concentrations of said excipients and the therapeutic agent are well known in the art and are described in standard textbooks. See, for example, “Amorphous Solid Dispersions Theory and Practice" by Navnit Shah et al.
In one aspect, the PD-1 antagonist is included in a pharmaceutical composition with a pharmaceutically acceptable carrier or diluent and may include additional pharmaceutically acceptable excipients.
Kits
The therapeutic agents of the combination therapies of the present invention may conveniently be combined in the form of a kit suitable for coadministration of the therapeutic agents.
In one aspect, the present invention relates to a kit comprising a first container, a second container, a third container, and a package insert, wherein the first container comprises at least one dose of encorafenib or a pharmaceutically acceptable salt thereof, the second container comprises at least one dose of cetuximab, the third container comprises at least one dose of pembrolizumab, and the package insert comprises instructions for treating a subject for cancer using the medicaments.
The kit may be particularly suitable for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist compliance, the kit typically includes directions for administration and may be provided with a memory aid. The kit may further comprise other materials that may be useful in administering the medicaments, such as diluents, filters, IV bags and lines, needles and syringes, and the like.
The following abbreviations are used throughout and in the Example below:
Figure imgf000051_0001
Figure imgf000052_0001
EXAMPLE 1
Phase 2 study of encorafenib and cetuximab plus pembrolizumab versus pembrolizumab alone in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC
RATIONALE AND OBJECTIVES
The purpose of this clinical study is to evaluate the safety and efficacy of encorafenib and cetuximab in combination with pembrolizumab compared with pembrolizumab alone in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC.
Primary Objective
To demonstrate that the combination of encorafenib and cetuximab and pembrolizumab is superior to pembrolizumab in prolonging progression-free survival (PFS), in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC.
Figure imgf000053_0001
• To demonstrate that the combination of encorafenib and cetuximab and pembrolizumab is superior to pembrolizumab in prolonging OS, in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC.
• To demonstrate that the combination of encorafenib and cetuximab and pembrolizumab is superior to pembrolizumab in prolonging objective response in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC.
• To demonstrate that the combination of encorafenib and cetuximab and pembrolizumab is superior to pembrolizumab in prolonging DOR in subjects with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC.
• To evaluate the overall safety and tolerability of encorafenib and cetuximab and pembrolizumab administered in combination compared to administration of pembrolizumab alone.
• To confirm the BRAF and MSI status in tumor tissue.
• To evaluate the effect on PROs in subjects treated with the combination of encorafenib and cetuximab and pembrolizumab compared to subjects treated with pembrolizumab alone with respect to the following: o EORTC QLQ-C30: change from baseline in the global health status/QoL, functional and symptom scales, and single items o EQ-5D-5L: change from baseline in the index score and VAS o PGIS score: change from baseline in the score o PGIC score
METHODS
Clinical Trial Design:
This is an open-label, multi-center, randomized Phase 2 study of encorafenib and cetuximab plus pembrolizumab (Triplet Arm [Arm A]) versus pembrolizumab alone (Control Arm [Arm B]) in participants with previously untreated BRAF V600E-mutant and MSI-H/dMMR mCRC. The primary objective of the study is to compare the efficacy, as measured by the primary endpoint of PFS per investigator, of Arm A versus Arm B.
Treatment:
Subject will be randomized in a 1 :1 ratio to Arm A or Arm B. Randomization will be stratified by ECOG (0 vs 1 ). Participants in Arm B are not eligible for crossover to Arm A. Participants will receive either:
• encorafenib (300 mg QD orally) + cetuximab (500 mg/m2 Q2W IV) + pembrolizumab (400 mg Q6W IV) (Triplet Arm [Arm A]) or
• pembrolizumab (400 mg Q6W IV) (Control Arm [Arm B]).
Study intervention will be administered until progressive disease (PD) per RECIST v1 .1 or until any of the other protocol-defined criteria for discontinuation of study intervention are met, whichever occurs first. In certain circumstances, continuation of treatment beyond PD may be allowed. In both treatment arms, duration of pembrolizumab treatment will not exceed 18 administrations (~24 months); and in Arm A, treatment with encorafenib and cetuximab will continue beyond ~24 months until PD per RECIST v1 .1 or until any of the other protocol-defined criteria for discontinuation of study intervention are met, whichever occurs first. After discontinuation of all study intervention, participants will be followed for safety, disease status, subsequent anticancer therapy, and survival status until withdrawal of consent/assent, the participant is lost to follow-up, death, or defined end of study, whichever occurs first.
Dose modifications
No dose adjustments for pembrolizumab is recommended. Pembrolizumab will be withheld for treatment-related toxicities as appropriate. Participants should receive appropriate supportive care measures deemed necessary. The dose of pembrolizumab will remain constant at [400 mg Q6W] for each dose level for encorafenib and cetuximab and in each arm.
The recommended dose reductions for encorafenib and cetuximab are presented in the following table. Dose reductions beyond the second dose reduction are not allowed for encorafenib and cetuximab.
Figure imgf000054_0001
Inclusion Criteria:
Participants are eligible to be included in the study only if all of the following criteria apply:
1 . Locally confirmed dMMR or MSI-H disease in tumor tissue or blood (e.g., ctDNA genetic testing) as determined by a local laboratory assay in a CLIA- or similarly certified laboratory.
2. Locally confirmed BRAF V600E mutation in tumor tissue or blood (e.g., circulating tumor DNA (ctDNA) genetic testing) as determined by either PCR or NGS-based local laboratory assay in a CLIA- or similarly certified laboratory.
3. Male or female participants age >16 years at the time of informed consent/assent (or the minimum country specific age of consent if >16). In countries or sites where enrollment of adolescents is not permitted (e.g., Germany), male or female participants age >18 years at the time of informed consent.
4. Participants who are willing and able to comply with all scheduled visits, treatment plan, laboratory tests, lifestyle considerations, and other study procedures.
5. Histologically or cytolog ically confirmed metastatic Stage IV colorectal adenocarcinoma. Note: Patients with oligometastatic disease previously treated with curative intent are eligible to participate in the study as long as they have baseline measurable disease per RECIST 1.1.
6. Presence of measurable disease per RECIST v1 .1 , as assessed by investigator and evidenced by available baseline tumor scan. Note: Baseline scan is defined as the last scan prior to the date of randomization. Note: Baseline scans will be required to be available for subsequent submission to a central radiology vendor to be assessed by the BICR.
7. Availability of adequate tumor tissue (primary or metastatic; archival or newly obtained; block or slides). Whenever possible, the archival sample should be from the same tumor block that was used for local BRAF V600E mutation and MSI-H/dMMR testing. This tissue block should be obtained from a biopsy or surgery that was performed within 2 years prior to study enrollment. A newly obtained tumor tissue biopsy must be provided prior to randomization for participants unable to provide adequate archival tumor tissue. If a newly obtained biopsy is taken, the biopsy should be taken from a nontarget lesion when possible.
8. Have not received prior systemic regimens for metastatic disease. Note: Participants with early-stage disease (e.g., Stages l-lll) treated with surgery followed by chemotherapy (e.g., treatment in the adjuvant setting) or have received prior systemic neoadjuvant therapy with or without radiation who present with new lesions or evidence of disease recurrence during or within 6 months of the last dose of chemotherapy would be considered as having received 1 prior systemic therapy in the metastatic setting.
9. ECOG performance status of <1 .
10. Adequate bone marrow function characterized by the following at screening: a. ANC >1.5 x 109/L b. Platelets >100 x 109/L c. Hemoglobin >9.0 g/dL (without blood transfusions 2 weeks prior to randomization)
1 1 . Adequate hepatic and renal function characterized by the following at screening: a. Serum Tbili <1 .5 x upper limit of normal (ULN) and < 2 mg/dL. Note: Tbili >1 .5 x ULN is allowed if direct (conjugated) < 1.5 x ULN and indirect (unconjugated) bilirubin is < 4.25 x ULN. Note: Participants with documented Gilbert syndrome or hyperbilirubinemia due to non-hepatic cause (e.g., hemolysis, hematoma) may be enrolled following discussion and agreement with the sponsor or designee. b. alanine aminotransferase (ALT) and aspartate aminotransferase (AST) < 2.5 x ULN, or < 5 x ULN in the presence of liver metastases. c. Adequate renal function defined by an estimated creatinine clearance
Exclusion Criteria:
Participants are excluded from the study if any of the following criteria apply:
1. Colorectal adenocarcinoma that is RAS mutant or for which RAS mutation status is unknown.
2. Documented clinical disease progression (e.g., worsening of performance status, clinical symptoms, or clinically significant laboratory parameters demonstrating worsening of disease) or radiographic disease progression during the screening period.
3. Has active CNS metastases and/or carcinomatous meningitis. Participants with previously treated brain metastases may participate provided they are radiologically stable, i.e., without evidence of progression for at least 4 weeks by repeat imaging (note that the repeat imaging should be performed during study screening), clinically stable and without requirement of steroid treatment for at least 14 days prior to first dose of study intervention.
4. Leptomeningeal disease.
5. Diagnosis of immunodeficiency or an active autoimmune disease that required systemic treatment in the past 2 years (i.e., with use of disease modifying agents, corticosteroids, or immunosuppressive drugs). Note: Participants with diabetes type I, vitiligo, psoriasis, controlled asthma, Graves’ disease, Hashimoto’s disease or hypo- or hyperthyroid disease are exceptions and may participate. Note: Replacement and symptomatic therapies (e.g., levothyroxine, insulin, or physiologic corticosteroid replacement therapy for adrenal or pituitary insufficiency) are not considered a form of immunosuppressive agents and are permitted.
6. Presence of acute or chronic pancreatitis. 7. History of chronic inflammatory bowel disease requiring medical intervention (immunomodulatory or immunosuppressive medications or surgery) < 12 months prior to randomization.
8. Unable to swallow, retain, and absorb oral medications.
9. Impaired gastrointestinal function (e.g., uncontrolled nausea, vomiting or diarrhea, malabsorption syndrome, small bowel resection) or disease which may significantly alter the absorption of oral study intervention or recent changes in bowel function suggesting current or impending bowel obstruction.
10. Clinically significant cardiovascular diseases, including any of the following: a. History of acute myocardial infarction, acute coronary syndromes (including unstable angina, coronary artery bypass graft, coronary angioplasty or stenting) < 6 months prior to randomization. b. Congestive heart failure requiring treatment (New York Heart Association Grade > 2). c. Recent history (one year) or presence of clinically significant cardiac arrhythmias (including uncontrolled atrial fibrillation or uncontrolled paroxysmal supraventricular tachycardia). d. History of thromboembolic or cerebrovascular events < 12 weeks prior to randomization. Examples include transient ischemic attacks, cerebrovascular accidents, hemodynamically significant (i.e., massive or sub-massive) deep vein thrombosis or pulmonary emboli. Note: Participants with either deep vein thrombosis or pulmonary emboli that do not result in hemodynamic instability are allowed to enroll as long as they are on a stable dose of anticoagulants for at least 4 weeks. Note: Participants with thromboembolic events related to indwelling catheters (including PICC lines) or other procedures may be enrolled. e. Triplicate average QTcF interval >480 ms or a history of prolonged QT syndrome. Note: Participants with BBB or with an implanted cardiac pacemaker may enroll into the study upon agreement between the investigator and sponsor or designee. f. LQTS.
11. Has a history of (non-infectious) pneumonitis / interstitial lung disease that required steroids or has current pneumonitis / interstitial lung disease.
12. Evidence of active and uncontrolled bacterial or viral infection, with certain exceptions, as noted below, for chronic infection with hepatitis B or hepatitis C, within 2 weeks prior to start of study intervention. Note: For COVID-19/SARS-CoV-2, SARS-CoV-2 testing is not mandated for study entry, and testing should follow local clinical practice standards. Any participant with a positive test result for SARS-CoV-2 infection, is known to have asymptomatic infection or is suspected of having SARS-CoV-2, is excluded. Once the infection resolves, the participant may be considered for re-screening.
13. Participants positive for HIV are ineligible unless they meet all of the following: a. A stable regimen of highly active anti-retroviral therapy that is not contraindicated. b. No requirement for concurrent antibiotics or antifungal agents for the prevention of opportunistic infections c. A CD4 count >250 cells/mcL, and an undetectable HIV viral load on standard PCR- based tests.
Note: Participants with a history of Kaposi sarcoma and/or Multicentric Castleman Disease are not eligible.
14. Active hepatitis B or hepatitis C infection a. Active HBV is defined as any of the following:
• HBsAg(+), HBV DNA >200 lU/mL (105 copies/mL).
• HBsAg(+), HBV DNA <200 ILI/mL and persistent or intermittent elevation of ALT/AST and/or liver biopsy showing chronic hepatitis with moderate or severe necroinflammation.
Note: Participants who are HBsAg(-), HBcAb(+) are eligible and should be monitored/treated as per local standard of care. b. Active HCV is defined as:
• HCV antibody positive; AND
• Presence of HCV RNA
15. Concurrent or previous other malignancy within 2 years of study entry, except curatively treated basal or squamous cell skin cancer, prostate intraepithelial neoplasm, carcinoma in-situ of the cervix, Bowen’s disease or prostate cancer with a Gleason score < 6. Participants with a history of other curatively treated malignancies with low risk of recurrence not listed may also be considered eligible after consultation with sponsor or designee.
16. Residual CTCAE > Grade 2 toxicity from any prior anticancer therapy, with the exception of Grade 2 alopecia. Participants with < Grade 2 neuropathy may be eligible.
17. Other medical or psychiatric condition including recent (within the past year) or active suicidal ideation/behavior or laboratory abnormality that may increase the risk of study participation or, in the investigator’s judgment, make the participant inappropriate for the study. Primary Endpoint:
• PFS per investigator, defined as the time from randomization until PD based on investigator assessment per RECIST v1 .1 or death due to any cause, whichever occurs first.
Secondary Endpoints:
• Incidence and severity of adverse events (AEs) graded according to the NCI CTCAE v4.03 and changes in clinical laboratory test parameters, vital signs and ECGs
• Incidence of dosing interruptions, dose modifications and permanent discontinuations associated with AEs
• OS, defined as the time from the date of randomization to the date of death due to any cause
• Objective response, defined as confirmed CR or confirmed PR based on investigator assessment per RECIST v1 .1 , from the date of randomization until the date of the first documentation of PD, death or start of new anticancer therapy
• DOR, defined as the time from the first response, until PD based on investigator assessment per RECIST v1 .1 or death due to any cause, whichever occurs first
• BRAF and MSI-status as determined by retrospective central testing of baseline tumor tissue
• Effect on PROs with respect to the following: o EORTC QLQ-C30: change from baseline in the global health status/QoL, functional and symptom scales, and single items o EQ-5D-5L: change from baseline in the index score and VAS o Patient Global Impression of Severity (PGIS) score: change from baseline in the score o Patient Global Impression of Change (PGIC) score
All publications and patent applications cited in the specification are herein incorporated by reference in their entirety. Although the foregoing invention has been described in some detail byway of illustration and example, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

We claim:
1. A method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
2. A method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
3. A method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
4. A method of treating previously untreated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising: a) detecting or having detected a BRAF V600E mutation from a biopsy of the cancer or a peripheral blood sample from the subject; b) identifying or having identified one or both of dMMR status from a biopsy of the cancer from the subject, and MSI-H status from a biopsy of the cancer or a peripheral blood sample from the subject; and c) administering to the subject a combination therapy consisting essentially of an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
5. The method according to any one of claims 1 to 4, wherein the encorafenib or pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 150 mg or about 225 mg or about 300 mg calculated as the free base equivalent.
6. The method according to any one of claims 1 to 4, wherein the encorafenib or pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 150 mg calculated as the free base equivalent.
7. The method according to any one of claims 1 to 4, wherein the encorafenib or pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 225 mg calculated as the free base equivalent.
8. The method according to any one of claims 1 to 4, wherein the encorafenib or pharmaceutically acceptable salt thereof is administered orally as a once daily dosage of about 300 mg calculated as the free base equivalent.
9. The method according to any one of claims 1 to 8, wherein the encorafenib is formulated as a capsule.
10. The method according to claim 9, wherein said capsule comprises 50 mg of encorafenib calcuated as the free base.
1 1. The method according to claim 9, wherein said capsule comprises 75 mg of encorafenib calcuated as the free base.
12. The method according to any one of claims 1 to 11 , wherein the encorafenib is a free base.
13. The method according to any one of claims 1 to 12, wherein the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 300 mg/m2 or about 400 mg/m2 or about 500 mg/m2.
14. The method according to any one of claims 1 to 12, wherein the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 300 mg/m2
15. The method according to any one of claims 1 to 12, wherein the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 400 mg/m2.
16. The method according to any one of claims 1 to 12, wherein the cetuximab is administered as an intravenous infusion every two weeks at a dose of about 500 mg/m2.
17. The method according to any one of claims 1 to 16, wherein the pembrolizumab is administered every 6 weeks at a dose of about 400 mg.
18. The method according to any one of claims 1 to 17, wherein the amounts together are effective in prolonging progression-free survival as compared to treatment with pembrolizumab alone.
19. The method according to any one of claims 1 to 18, wherein the amounts together are effective in prolonging overall survival as compared to treatment with pembrolizumab alone.
20. The method according to any one of claims 1 to 19, wherein the amounts together are effective in prolonging objective response as compared to treatment with pembrolizumab alone.
21 . The method according to any one of claims 1 to 20, wherein the amounts together are effective in prolonging duration of response as compared to treatment with pembrolizumab alone.
22. The method according to any one of claims 1 to 21 , wherein the colorectal cancer is metastatic or unresectable colorectal cancer.
23. The method according to any one of claims 1 to 22, wherein the colorectal cancer is BRAF V600E-mutant, MSI-H colorectal cancer.
24. The method according to any one of claims 1 to 22, wherein the colorectal cancer is BRAF V600E-mutant, dMMR colorectal cancer.
25. The method according to any one of claims 1 to 22, wherein the colorectal cancer is BRAF V600E-mutant, MSI-H, dMMR colorectal cancer.
26. The method according to any one of claims 1 to 25, wherein the subject is a human.
27. The method according to claim 16, wherein the pembrolizumab is administered as an intravenous infusion.
28. The method according to any one of claims 1 to 16, wherein the pembrolizumab is administered every 3 weeks at a dose of about 200 mg.
29. A method of treating previously treated BRAF V600E-mutant, MSI-H/dMMR colorectal cancer in a subject in need thereof, comprising administering to the subject a combination therapy comprising an amount of encorafenib, or a pharmaceutically acceptable salt thereof, an amount of cetuximab, and an amount of pembrolizumab, wherein the amounts together are effective in treating said cancer.
PCT/US2023/067469 2022-05-25 2023-05-25 Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer WO2023230554A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263345815P 2022-05-25 2022-05-25
US63/345,815 2022-05-25
US202363461393P 2023-04-24 2023-04-24
US63/461,393 2023-04-24

Publications (1)

Publication Number Publication Date
WO2023230554A1 true WO2023230554A1 (en) 2023-11-30

Family

ID=87036721

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/067469 WO2023230554A1 (en) 2022-05-25 2023-05-25 Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer

Country Status (1)

Country Link
WO (1) WO2023230554A1 (en)

Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
US7332582B2 (en) 2002-05-23 2008-02-19 Curetech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2011025927A1 (en) 2009-08-28 2011-03-03 Irm Llc Compounds and compositions as protein kinase inhibitors
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
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
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2013070996A1 (en) 2011-11-11 2013-05-16 Novartis Ag Method of treating a proliferative disease
US8609089B2 (en) 2008-08-25 2013-12-17 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
WO2014018725A1 (en) 2012-07-26 2014-01-30 Novartis Ag Pharmaceutical combinations of a cdk4/6 inhibitor and a b-raf inhibitor
WO2014025688A1 (en) 2012-08-07 2014-02-13 Novartis Ag Pharmaceutical combinations comprising a b-raf inhibitor, an egfr inhibitor and optionally a pi3k-alpha inhibitor
WO2014072493A1 (en) 2012-11-08 2014-05-15 Novartis Ag Pharmaceutical combination comprising a b-raf inhibitor and a histone deacetylase inhibitor and their use in the treatment of proliferative diseases
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2014147573A2 (en) 2013-03-21 2014-09-25 Novartis Ag Combination therapy
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2014194302A2 (en) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Antigen binding proteins that bind pd-1
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2017210538A1 (en) 2016-06-03 2017-12-07 Giordano Caponigro Pharmaceutical combinations
WO2020055992A1 (en) 2018-09-11 2020-03-19 NetraDyne, Inc. Inward/outward vehicle monitoring for remote reporting and in-cab warning enhancements
WO2020261156A1 (en) 2019-06-28 2020-12-30 Array Biopharma Inc. Quinazolin-4-one derivatives useful for the treatment of braf-associated diseases and disorders
WO2021250521A1 (en) 2020-06-09 2021-12-16 Array Biopharma Inc. 4-oxo-3,4-dihydroquinazolinon compounds for the treatment of braf-associated diseases and disorders

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US7332582B2 (en) 2002-05-23 2008-02-19 Curetech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human 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
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US8609089B2 (en) 2008-08-25 2013-12-17 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
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
WO2011025927A1 (en) 2009-08-28 2011-03-03 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2013070996A1 (en) 2011-11-11 2013-05-16 Novartis Ag Method of treating a proliferative disease
WO2014018725A1 (en) 2012-07-26 2014-01-30 Novartis Ag Pharmaceutical combinations of a cdk4/6 inhibitor and a b-raf inhibitor
WO2014025688A1 (en) 2012-08-07 2014-02-13 Novartis Ag Pharmaceutical combinations comprising a b-raf inhibitor, an egfr inhibitor and optionally a pi3k-alpha inhibitor
WO2014072493A1 (en) 2012-11-08 2014-05-15 Novartis Ag Pharmaceutical combination comprising a b-raf inhibitor and a histone deacetylase inhibitor and their use in the treatment of proliferative diseases
WO2014147573A2 (en) 2013-03-21 2014-09-25 Novartis Ag Combination therapy
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2014194302A2 (en) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Antigen binding proteins that bind pd-1
US20150079109A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
WO2017210538A1 (en) 2016-06-03 2017-12-07 Giordano Caponigro Pharmaceutical combinations
WO2020055992A1 (en) 2018-09-11 2020-03-19 NetraDyne, Inc. Inward/outward vehicle monitoring for remote reporting and in-cab warning enhancements
WO2020261156A1 (en) 2019-06-28 2020-12-30 Array Biopharma Inc. Quinazolin-4-one derivatives useful for the treatment of braf-associated diseases and disorders
WO2021250521A1 (en) 2020-06-09 2021-12-16 Array Biopharma Inc. 4-oxo-3,4-dihydroquinazolinon compounds for the treatment of braf-associated diseases and disorders

Non-Patent Citations (44)

* Cited by examiner, † Cited by third party
Title
"Handbook of Pharmaceutical Excipients", 1999, AMERICAN PHARMACEUTICAL ASSOCIATION
"Information for KEYTRUDA", 2014, MERCK & CO., INC.
"NCBI", Database accession no. NP005009.
"Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
"Remington, The Science and Practice of Pharmacy", 2000, MACK PUBLISHING
AHMADZADEH, MOJGAN ET AL.: "Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired", BLOOD, vol. 114, 2009, pages 1537 - 1544
ANDRE ET AL., N. ENGL. J. MED., vol. 383, no. 23, 2020, pages 2207 - 2218
ANGAL ET AL., MOL. IMUNOL., vol. 30, 1993, pages 105
ANONYMOUS: "A PHASE 2, RANDOMIZED, OPEN-LABEL STUDY OF ENCORAFENIB AND CETUXIMAB PLUS PEMBROLIZUMAB VERSUS PEMBROLIZUMAB ALONE IN PARTICIPANTS WITH PREVIOUSLY UNTREATED BRAF V600E-MUTANT, MSI H/DMMR METASTATIC COLORECTAL CANCER", CLINICALTRIALS.GOV, 17 March 2022 (2022-03-17), XP093077970, Retrieved from the Internet <URL:https://clinicaltrials.gov/study/NCT05217446?tab=history&a=2> [retrieved on 20230901] *
B. C. FINNINT. M. MORGAN, J. PHARM. SCI., vol. 88, 1999, pages 955 - 958
CHANG, H.I.YEH, M.K.: "Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy", INT J NANOMEDICINE, vol. 7, 2012, pages 49 - 60, XP055069345, DOI: 10.2147/IJN.S26766
CHOTHIA ET AL., J MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 878 - 883
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
DONG, HAIDONG ET AL.: "Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion", NAT MED., vol. 8, no. 8, August 2002 (2002-08-01), pages 793 - 800, XP002397368, DOI: 10.1038/nm730
GAO, QIANG ET AL.: "Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma", CLINICAL CANCER RESEARCH, vol. 15, 2009, pages 971 - 979
GHEBEH, HAZEM ET AL.: "Foxp3+ tregs and B7-H1+/PD-1+ T lymphocytes co-infiltrate the tumor tissues of high-risk breast cancer patients: implication for immunotherapy", BMC CANCER, vol. 8, 23 February 2008 (2008-02-23), pages 57, XP021034690
GHEBEH, HAZEM ET AL.: "The B7-H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors", NEOPLASIA, vol. 8, 2006, pages 190 - 198, XP008091677, DOI: 10.1593/neo.05733
HAMANISHI, JUNZO ET AL.: "Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer", PROC. NATL. ACAD. SCI. USA, vol. 104, 2007, pages 3360 - 3365
HINO ET AL.: "Tumor cell expression of programmed cell death-1 is a prognostic factor for malignant melanoma", CANCER, vol. 00, 2010, pages 1 - 9
HOOVER, JOHN E.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
INMAN, BRANT A ET AL.: "PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression", CANCER, vol. 109, 2007, pages 1499 - 1505
KABAT ET AL., J. BIOL. CHEM., vol. 252, 1977, pages 6609 - 6616
KABAT, ADV. PROT. CHEM., vol. 32, 1978, pages 1 - 75
KARKI SOPHIYA ET AL: "Treating Colorectal Cancer with Immunotherapy: Implications for Single Versus Combination Therapy", CURRENT COLORECTAL CANCER REPORTS, SPRINGER US, BOSTON, vol. 16, no. 5, 20 August 2020 (2020-08-20), pages 107 - 117, XP037234149, ISSN: 1556-3790, [retrieved on 20200820], DOI: 10.1007/S11888-020-00459-Y *
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
NAKANISHI, JURO ET AL.: "Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers", CANCER IMMUNOL IMMUNOTHER., vol. 56, 2007, pages 1173 - 1182, XP019514169, DOI: 10.1007/s00262-006-0266-z
NAVNIT SHAH, AMORPHOUS SOLID DISPERSIONS THEORY AND PRACTICE
NOMI, TAKEO ET AL.: "Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer", CLINICAL CANCER RESEARCH, vol. 13, 2007, pages 2151 - 2157, XP002533527, DOI: 10.1158/1078-0432.CCR-06-2746
OHIGASHI, YUICHIRO ET AL.: "Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand 2 expression in human esophageal cancer", CLIN. CANCER RESEARCH, vol. 11, 2005, pages 2947 - 2953, XP002419631, DOI: 10.1158/1078-0432.CCR-04-1469
PRESTA, J. ALLERGY CLIN. IMMUNOL., vol. 116, 2005, pages 731
RODRIQUENZ MARIA GRAZIA ET AL: "Exploring biological heterogeneity and implications on novel treatment paradigm in BRAF-mutant metastatic colorectal cancer", CRITICAL REVIEWS IN ONCOLOGY/HEMATOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 173, 23 March 2022 (2022-03-23), XP087033327, ISSN: 1040-8428, [retrieved on 20220323], DOI: 10.1016/J.CRITREVONC.2022.103657 *
ROGERS JANE E. ET AL: "Pharmacotherapy for unresectable metastatic colorectal cancer", EXPERT OPIN PHARMACOTHER, vol. 23, no. 2, 26 September 2021 (2021-09-26), London, UK, pages 211 - 220, XP093077858, ISSN: 1465-6566, DOI: 10.1080/14656566.2021.1982895 *
SCHUURMAN, J, MOL. IMMUNOL., vol. 38, 2001, pages 1 - 8
SHARPE, ARLENE H ET AL.: "The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection", NATURE IMMUNOLOGY, vol. 8, 2007, pages 239 - 245
SHIMAUCHI, TAKATOSHI ET AL.: "Augmented expression of programmed death-1 in both neoplasmatic and nonneoplastic CD4+ T-cells in adult T-cell Leukemia/ Lymphoma", INT. J. CANCER, vol. 121, 2007, pages 2585 - 2590
THOMPSON, R HOUSTON ET AL.: "PD-1 is expressed by tumor infiltrating cells and is associated with poor outcome for patients with renal carcinoma", CLINICAL CANCER RESEARCH, vol. 15, 2007, pages 1757 - 1761, XP055613062, DOI: 10.1158/1078-0432.CCR-06-2599
THOMPSON, R HOUSTONEUGENE D KWON: "Significance of B7-H1 overexpression in kidney cancer", CLINICAL GENITOURIN CANCER, vol. 5, 2006, pages 206 - 211, XP009114210, DOI: 10.3816/CGC.2006.n.038
VENDERBOSCH ET AL., CLINICAL CANCER RESEARCH, vol. 20, no. 20, 2014, pages 5322 - 5330
W. A. WEBER, J. NUCL. MED., vol. 50, no. 200, pages 1S - 10S
WATSON ET AL.: "Molecular Biology of the Gene", 1987, THE BENJAMIN/CUMMINGS PUB. CO., pages: 224
WHO DRUG INFORMATION, vol. 27, no. 1, 2013, pages 161 - 162
YANG, WANHUA ET AL.: "PD-1 interaction contributes to the functional suppression of T-cell responses to human uveal melanoma cells in vitro", INVEST OPHTHALMOL VIS SCI., vol. 49, no. 6, 2008, XP055412241, DOI: 10.1167/iovs.07-1606

Similar Documents

Publication Publication Date Title
CN105263520B (en) Compositions comprising anti-CD 38 antibodies and lenalidomide
US20230181605A1 (en) Triple combination therapy for enhancing cancer cell killing in cancers with low immunogenicity
JP2018507220A (en) Methods, compositions and kits for the treatment of cancer
KR20210120022A (en) Composite pharmaceutical composition for tumor treatment
JP2021054832A (en) Pharmaceutical combinations
EP3939610A1 (en) Combined pharmaceutical composition for treating small cell lung cancer
JP2021130668A (en) Treatment for multiple myeloma (mm)
WO2020233602A1 (en) Quinoline derivative used for combination treatment of small cell lung cancer
CN112043831A (en) Quinolines for use in the combined treatment of breast cancer
EP4327822A1 (en) Use of anti-pd-1 antibody in combination with first-line chemotherapy for treating advanced non-small cell lung cancer
US20210355224A1 (en) Methods and combinations for the treatment of cancer using immune checkpoint inhibitor antibodies
WO2023230554A1 (en) Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer
CN113473989B (en) Administration of SUMO activating enzyme inhibitors and checkpoint inhibitors
JP2023539506A (en) Use of anti-PD-1 antibodies and cytotoxic anticancer drugs in the treatment of non-small cell lung cancer
CN113117072A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
JP2022515371A (en) Treatment of Breast Cancer with Combination Therapy Containing AKT Inhibitors, Taxanes and PD-L1 Inhibitors
KR20240028452A (en) Methods and compositions for treating cancer
KR20240038008A (en) Cancer treatment methods and compositions
JP2024513505A (en) Compositions and methods for treating tumors
WO2024052514A1 (en) Compositions and methods for treating advanced solid tumors
TW202342527A (en) Treatment of squamous non small cell lung cancer
TW202315631A (en) Trimodality therapy to enhance cancer cell killing in poorly-immunogenic cancers can induce immunogens and inhibit immune checkpoint
KR20240014052A (en) Methods of allowing intratumoral infiltration of immune cells
CN116761610A (en) AHR inhibitors and uses thereof

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: 23734855

Country of ref document: EP

Kind code of ref document: A1