CN117858725A - ERK1/2 and EGFR inhibitor combination therapies - Google Patents

ERK1/2 and EGFR inhibitor combination therapies Download PDF

Info

Publication number
CN117858725A
CN117858725A CN202280057485.0A CN202280057485A CN117858725A CN 117858725 A CN117858725 A CN 117858725A CN 202280057485 A CN202280057485 A CN 202280057485A CN 117858725 A CN117858725 A CN 117858725A
Authority
CN
China
Prior art keywords
cancer
day
inhibitor
administered
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280057485.0A
Other languages
Chinese (zh)
Inventor
罗伯特·菲尔德·舒梅克
艾琳·丹尼斯·卢
林卫
莱斯利·哈里斯·布雷尔
莱努斯·马丁
张京川
乔安妮·吴
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Yirui Shikang Pharmaceutical Research And Development Co
Original Assignee
Yirui Shikang Pharmaceutical Research And Development Co
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 Yirui Shikang Pharmaceutical Research And Development Co filed Critical Yirui Shikang Pharmaceutical Research And Development Co
Priority claimed from PCT/US2022/034660 external-priority patent/WO2022271907A1/en
Publication of CN117858725A publication Critical patent/CN117858725A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure relates generally to the use of ERK1/2 inhibitors in combination with EGFR inhibitors for the treatment of cancer, in particular solid tumors. The disclosure also relates to the use of an ERK1/2 inhibitor in combination with an EGFR inhibitor and a BRAF inhibitor for the treatment of cancer, in particular solid tumors.

Description

ERK1/2 and EGFR inhibitor combination therapies
Cross reference
The present application claims the benefit of U.S. provisional application Ser. No. 63/214,765, U.S. provisional application Ser. No. 63/236,635, U.S. provisional application Ser. No. 63/277,547, U.S. provisional application Ser. No. 63/279,877, U.S. provisional application Ser. No. 63/321,605, U.S. provisional application Ser. No. 63/877, and U.S. provisional application Ser. No. 63/321,605, U.S. provisional application Ser. No. 63/877, U.S. provisional application Ser. No. 3/2022, U.S. Ser. No. 63/321,605, and U.S. No. 3/18, 2022, each of which is incorporated herein by reference in its entirety.
Background
ERK1 and ERK2 (collectively, "ERK 1/2") are related protein-serine/threonine kinases that are involved in the Ras-Raf-MEK-ERK signal transduction pathway, and the like, sometimes referred to as the mitogen-activated protein kinase (MAPK) pathway. This pathway is thought to play an important role in regulating many basic cellular processes including one or more of cell proliferation, survival, adhesion, cyclical progression, migration, differentiation, metabolism and transcription. Activation of the MAPK pathway has been reported in many tumor types, including lung, colon, pancreas, kidney, and ovary. Thus, substances that can reduce activation may be of interest for possible treatments.
Disclosure of Invention
MEK appears to activate ERK1/2 through phosphorylation of threonine and tyrosine residues (i.e., at Tyr204/187 and Thr 202/185). Once activated, ERK1/2 catalyzes the phosphorylation of serine/threonine residues of over 100 substrates, and activates both cytoplasmic and nuclear proteins associated with cell growth, proliferation, survival, angiogenesis, and differentiation (all markers of cancer phenotypes). Thus, it may be beneficial to target ERK1 and ERK 2 to develop and use ERK1/2 inhibitors as a way to inhibit tumor growth.
In addition, ERK inhibitors may have utility in combination with other kinase (e.g., MAPK) inhibitors. Recently, researchers have reported that small molecule inhibitors have a synergistic effect on the dual inhibition of MEK and ERK and act to overcome acquired resistance to MEK inhibitors. See Hatzivessiliou et al ERK Inhibition Overcomes Acquired Resistance to MEK Inhibition mol. Cancer Ther.2012,11,1143-1154.
In addition to ERK1/2, the Epidermal Growth Factor Receptor (EGFR), a receptor that is a member of the epidermal growth factor family of extracellular protein ligands, also acts upstream of the RAS pathway.
EGFR is a Receptor Tyrosine Kinase (RTK), a protein that is embedded in the cell membrane and transmits growth signals from the external environment to intracellular mechanisms. In the resting state, this protein resides as an inactive monomer on the cell membrane. Growth factors secreted by nearby cells bind to specific RTKs, such as the growth factor EGF binds to EGFR, and cause dimerization of these RTKs. Dimerized RTKs are activated by transphosphorylation of their intracellular regions. Intracellular proteins, such as adaptor proteins, bind to these phosphorylated regions and propagate growth-promoting signals within the cell via one or more signaling pathways. The cells express a variety of RTKs such that environmental cues can be transmitted to specific cell populations in specific environments. EGFR mediates growth-promoting signaling in the skin and in the ducts and external surfaces of many organs. Overactive RTK signaling can lead to uncontrolled cell growth and survival, thereby converting normal cells into cancer cells.
The opportunity to target the signaling pathway from multiple angles and potentially improve the Ras upstream feedback loop via ERKl/2 and EGFR provides an opportunity to develop methods employing combination therapies.
Embodiments disclosed herein relate generally to compositions and methods related to combination therapies that utilize an ERK1/2 inhibitor in combination with an EGFR inhibitor to treat cancer while providing an unexpected degree of synergy.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of (i) compound 1:or a pharmaceutically acceptable salt thereof, and
(ii) EGFR inhibitors.
In some embodiments, the EGFR inhibitor is a small molecule EGFR inhibitor.
In some embodiments, the EGFR inhibitor is afatinib (afatinib), efonamab (amivantiamab), canetinib (canertinib), cetuximab (cetuximab), dacominib (dacominib), daphnetin (daphnetin), erlotinib (erlotinib), gefitinib (gefitinib), icotinib (icotinib), lapatinib (lapatinib), lasatinib (lazertinib), lifafinib (lifirafenanib), crizotuzumab (mirzotamab clezutoclax), mo Bo tinib (mobocetinib), natatinib (nazartinib), rituximab (dacominib), lenalitinib (neptinib), oxaltinib (osertinib), panitumumab (panitunib), litinib (petinib), 35, vanatinib (vantiltinib), vandulatinib (vantilapitinib), or vandulatinib (vantilapitinib).
In some embodiments, the EGFR inhibitor is octreotide.
In some embodiments, the octreotide is administered in an amount of about 80 mg/day.
In some embodiments, the EGFR inhibitor is cetuximab.
In some embodiments, cetuximab is initially at 400mg/m once a week 2 Applied over 120 minutes, then at 250mg/m 2 Applied over a period of 60 minutes.
In some embodiments, cetuximab is administered at 500mg/m once every two weeks 2 And (3) application.
In some embodiments, cetuximab is administered at 400mg/m once every two weeks 2 Administered or once every two weeks at 300mg/m 2 And (3) application.
In some embodiments, the method further comprises administering a BRAF inhibitor.
In some embodiments, the BRAF inhibitor is dabrafenib (dabrafenib), kang Naifei ni (encorafenib), regrafenib (regrafenib), sorafenib (sorafenib), or vemurafenib (vemurafenib).
In some embodiments, the BRAF inhibitor is Kang Naifei ni.
In some embodiments, kang Naifei Ni is administered in an amount of about 100 mg/day to about 500 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 450 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 300 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 225 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 150 mg/day.
In some embodiments, the BRAF inhibitor is dabrafenib.
In some embodiments, dabrafenib is administered in an amount of about 150 mg.
Also disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) An EGFR inhibitor; and
(iii) BRAF inhibitors.
In some embodiments, the EGFR inhibitor is a small molecule inhibitor.
In some embodiments, the EGFR inhibitor is afatinib, el Mo Tuoshan antibody, cetuximab, dactyltinib, erlotinib, gefitinib, lapatinib, lasatinib, lifenib, clomizotocet mab, mo Bo tinib, natatinib, cetuximab, lenatinib, octyitinib, or vandetanib.
In some embodiments, the EGFR inhibitor is octreotide.
In some embodiments, the octreotide is administered in an amount of about 80 mg/day.
In some embodiments, the EGFR inhibitor is cetuximab.
In some embodiments, cetuximab is initially at 400mg/m once a week 2 Applied over 120 minutes, then at 250mg/m 2 Applied over a period of 60 minutes.
In some embodiments, cetuximab is administered at 500mg/m once every two weeks 2 And (3) application.
In some embodiments, cetuximab is administered at 400mg/m once every two weeks 2 Administered or once every two weeks at 300mg/m 2 And (3) application.
In some embodiments, the BRAF inhibitor is dabrafenib, kang Naifei ni, regafinib, sorafenib, or vemurafenib.
In some embodiments, the BRAF inhibitor is Kang Naifei ni.
In some embodiments, kang Naifei Ni is administered in an amount of about 100 mg/day to about 500 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 450 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 300 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 225 mg/day.
In some embodiments, kang Naifei Ni is administered in an amount of about 150 mg/day.
In some embodiments, the BRAF inhibitor is dabrafenib.
In some embodiments, dabrafenib is administered in an amount of about 150 mg/day.
Also disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Cetuximab; and
(iii) Kang Naifei Ni.
A method of treating cancer in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Cetuximab; and
(iii) Darafenib.
Also disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Ornitinib; and
(iii) Kang Naifei Ni.
Also disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof
(ii) Ornitinib; and
(iii) Darafenib.
In some embodiments, the pharmaceutically acceptable salt of compound 1 is mandelate.
In some embodiments, the cancer is a mitogen-activated protein kinase (MAPK) pathway-driven cancer.
In some embodiments, the cancer is a BRAF-driven cancer, HRAS-driven cancer, or NRAS-driven cancer.
In some embodiments, the cancer comprises at least one cancer cell driven by deregulated ERK.
In some embodiments, the cancer has at least one mutation in the RAS. In some embodiments, the cancer has at least one mutation in RAF. In some embodiments, the cancer has at least one mutation in MEK.
In some embodiments, the cancer has a G12C KRAS mutation. In some embodiments, the cancer has a G12D KRAS mutation. In some embodiments, the cancer has a G12S KRAS mutation.
In some embodiments, the cancer has a G12V KRAS mutation. In some embodiments, the cancer has a G13D KRAS mutation. In some embodiments, the cancer has a Q16H KRAS mutation.
In some embodiments, the cancer has a Q16K KRAS mutation. In some embodiments, the cancer has a Q61R NRAS mutation.
In some embodiments, the cancer is a BRAF V600E or V600K mutant tumor.
In some embodiments, the cancer is a pan-carcinoma that does not receive MAPKm/MAPKi.
In some embodiments, the cancer comprises one or more EGFR mutations selected from the group consisting of: increased EGFR gene copy, EGFR gene amplification, chromosome 7 polyhedra, L858R, exon 19 deletion/insertion, L861Q, G719C, G719S, G719A, V765A, T783A, exon 20 insertion, EGFR splice variants (Viii, vvi and Vii), A289D, A289T, A289V, G598A, G598V, T790M and C797S.
In some embodiments, the cancer comprises one or more EGFR mutations selected from the group consisting of L858R, exon 19 deletion, and T790M.
In some embodiments, the cancer is a solid tumor.
In some embodiments, the cancer is non-small cell lung cancer (NSCLC), melanoma, pancreatic cancer, salivary gland tumor, thyroid cancer, colorectal cancer (CRC), or esophageal cancer.
In some embodiments, the cancer is non-small cell lung cancer (NSCLC). In some embodiments, the NSCLC is an EGFR mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12C mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12D mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12S mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12V mutant NSCLC. In some embodiments, the NSCLC is a KRAS G13D mutant NSCLC. In some embodiments, the NSCLC is a KRAS Q61H mutant NSCLC. In some embodiments, the NSCLC is a KRAS Q61K mutant NSCLC. In some embodiments, the NSCLC is an NRAS Q61R mutant NSCLC.
In some embodiments, the cancer is NSCLC that does not receive MAPKm/MAPKi.
In some embodiments, the cancer is BRAFi treated V600 NSCLC.
In some embodiments, the cancer is KRAS treated G12C NSCLC. In some embodiments, the cancer is KRAS treated G12D NSCLC. In some embodiments, the cancer is KRAS treated G12S NSCLC. In some embodiments, the cancer is KRAS treated G12V NSCLC. In some embodiments, the cancer is KRAS treated G13D NSCLC. In some embodiments, the cancer is KRAS treated Q61H NSCLC. In some embodiments, the cancer is KRAS treated Q61KNSCLC. In some embodiments, the cancer is NRAS treated Q61R NSCLC.
In some embodiments, the cancer is pancreatic cancer.
In some embodiments, the cancer is pancreatic cancer that does not receive MAPKm/MAPKi.
In some embodiments, the cancer is melanoma.
In some embodiments, the melanoma is a BRAF V600E or V600K mutant tumor.
In some embodiments, the cancer is BRAFi treated V600 melanoma.
In some embodiments, the cancer is a salivary gland tumor.
In some embodiments, the cancer is thyroid cancer.
In some embodiments, the cancer is colorectal cancer (CRC). In some embodiments, the CRC is a BRAF V600E CRC. In some embodiments, the CRC is a KRAS mutant CRC.
In some embodiments, the CRC is a KRAS G12C mutant CRC. In some embodiments, the CRC is a KRAS G12D mutant CRC. In some embodiments, the CRC is a KRAS G12S mutant CRC. In some embodiments, the CRC is a KRAS G12V mutant CRC. In some embodiments, the CRC is a KRAS G13D mutant CRC. In some embodiments, the CRC is a KRAS Q61H mutant CRC. In some embodiments, the CRC is a KRAS Q61K mutant CRC. In some embodiments, the CRC is an NRAS mutant CRC. In some embodiments, the CRC is an NRAS Q61R mutant CRC.
In some embodiments, the cancer is esophageal cancer.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day to about 300 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of 25 mg/day to 150 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day, about 50 mg/day, about 75 mg/day, about 100 mg/day, about 125 mg/day, about 150 mg/day, about 175 mg/day, about 200 mg/day, about 225 mg/day, or about 250 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day, about 50 mg/day, about 100 mg/day, or about 150 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 250 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered once daily (QD). In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered twice daily (BID). In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered three times per day (TID).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered once a week. In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered twice weekly.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 50mg, about 75mg, about 100mg, about 125mg, about 150mg, about 175mg, about 200mg, about 225mg, or about 250mg, twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 50mg, about 100mg, about 125mg, or about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 125mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered for at least one 28 day period.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered on days 1, 8, 15, and 22 of a 28 day cycle.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered on day 1, day 8, day 15 of the 28 day cycle.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered orally.
In some embodiments, the method further comprises administering an additional MAPK pathway inhibitor. In some embodiments, the additional MAPK pathway inhibitor is a KRAS inhibitor, NRAS inhibitor, HRAS inhibitor, PDGFRA inhibitor, PDGFRB inhibitor, MET inhibitor, FGFR inhibitor, ALK inhibitor, ROS1 inhibitor, TRKA inhibitor, TRKB inhibitor, TRKC inhibitor, EGFR inhibitor, IGFR1R inhibitor, GRB2 inhibitor, SOS inhibitor, ARAF inhibitor, BRAF inhibitor, RAF1 inhibitor, MEK2 inhibitor, c-Mycv, CDK4/6, inhibitor CDK2 inhibitor, FLT3 inhibitor, or ERK1/2 inhibitor. In some embodiments, the additional MAPK pathway inhibitor is a KRAS inhibitor. In some embodiments, the additional MAPK pathway inhibitor is a BRAF inhibitor. In some embodiments, the additional MAPK pathway inhibitor is an EGFR inhibitor. In some embodiments, the additional MAPK pathway inhibitor is CDK4/6. In some embodiments, the additional MAPK pathway inhibitor is a FLT3 inhibitor. In some embodiments, the additional MAPK pathway inhibitor is adaglazeb (adagaramib), afatinib, ASTX029, bimetainib, cobimatinib (cobimatinib), dactyltinib, erlotinib, gefitinib (gilitetinib), lapatinib, LTT462, LY3214996, rituximab, lenatinib, nimotuzumab (nimotuzumab), palbociclib (palbociclib), semmetinib (selumetinib), sotoracib (sotoraanib), trametinib (trametinib), ulitinib (ulixotinib), vandetanib, or vemurafenib. In some embodiments, the additional MAPK pathway inhibitor is adaglazeb. In some embodiments, the additional MAPK pathway inhibitor is gefitinib. In some embodiments, the additional MAPK pathway inhibitor is palbociclib. In some embodiments, the additional MAPK pathway inhibitor is panitumumab. In some embodiments, the additional MAPK pathway inhibitor is sotorubin.
Incorporation by reference
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
Drawings
The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosure are utilized, and the accompanying drawings of which:
figure 1A shows refractory BRAF at Kang Naifei ni/cetuximab V600E In vivo data for compound 1+ Kang Naifei ni and compound 1+ Kang Naifei ni/cetuximab in the RKO CDX model.
Fig. 1B shows refractory BRAF at Kang Naifei ni/cetuximab V600E In vivo data for compound 1+ Kang Naifei ni and compound 1+ Kang Naifei ni/cetuximab in the WiDr CDX model.
Fig. 2A shows in vivo data for compound 1+octenib in an octenib refractory EGFR mutant PDX model (LUN 2355-214).
FIG. 2B shows in vivo data for the compound 1+Oxitinib in an Oxitinib refractory EGFR mutant PDX model (LUN 2355-128-33).
Fig. 3A shows in vivo tumor growth curves for compound 1 alone, kang Naifei ni alone, and compound 1+ Kang Naifei ni in a Kang Naifei ni refractory BRAF V600E CR0004 PDX model.
Fig. 3B shows in vivo tumor growth curves for compound 1 alone, kang Naifei ni alone, cetuximab alone, compound 1+ Kang Naifei ni combination, compound 1+ Kang Naifei ni+cetuximab triple combination, and Kang Naifei ni+cetuximab combination in a Kang Naifei ni+cetuximab (EC) refractory BRAF V600ECR0004 PDX model.
Fig. 4A shows in vivo tumor growth curves for compound 1 alone, kang Naifei ni alone, and the combination of compounds 1+ Kang Naifei ni in the Kang Naifei ni refractory-BRAF V600E CRC1011 PDX model.
Fig. 4B shows in vivo tumor growth curves for compound 1 alone, kang Naifei ni alone, cetuximab alone, compound 1+ Kang Naifei ni combination, compound 1+ Kang Naifei ni+cetuximab triple combination, and Kang Naifei ni+cetuximab combination in a Kang Naifei ni+cetuximab (EC) refractory BRAF V600ECRC1011 PDX model.
Detailed Description
As used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an agent" includes a plurality of such agents, and reference to "the cell" includes reference to one or more cells (or cells) and equivalents thereof known to those skilled in the art, and so forth. When ranges are used herein for physical properties (e.g., molecular weight) or chemical properties (e.g., formula), it is intended to include all combinations and subcombinations of the ranges and specific embodiments therein. When referring to a number or range of values, the term "about" means that the number or range of values referred to is an approximation within experimental variability (or within statistical experimental error), and thus in some cases the number or range of values will vary from 1% to 15% of the number or range of values. The term "comprising" (and related terms such as "having" or "including") is not intended to exclude "consisting of" or "consisting essentially of" any of the other embodiments, e.g., embodiments of any of the compositions of matter, compositions, methods, or processes described herein, etc.
As used in the specification and the appended claims, the following terms have the meanings indicated below, unless otherwise indicated.
As used herein, the term "therapeutic agent" refers to an agent for treating, combating, ameliorating, preventing or ameliorating an undesired condition or disease in a patient. In some embodiments, a therapeutic agent such as compound 1 is involved in the treatment and/or amelioration of cancer.
When used in combination with a therapeutic agent, "administering" refers to the systemic or local administration of the therapeutic agent directly into or onto the target tissue, or to the patient, whereby the therapeutic agent positively affects the tissue to which it is targeted. Thus, as used herein, the term "administering" when used in conjunction with the compositions described herein can include, but is not limited to, providing the composition into or onto a target tissue; the composition is provided systemically to the patient, for example, by oral administration, whereby the therapeutic agent reaches the target tissue or cells. "administration" of the composition may be accomplished by injection, topical administration, and oral administration, or by other methods alone or in combination with other known techniques.
The term "animal" as used herein includes, but is not limited to, human and non-human vertebrates such as wild, domestic and farm animals. As used herein, the terms "patient," "subject," and "individual" are intended to include living organisms in which certain conditions as described herein may occur. Examples include humans, monkeys, cows, sheep, goats, dogs, cats, mice, rats and transgenic species thereof. In a preferred embodiment, the patient is a primate. In certain embodiments, the primate or subject is a human. In some cases, the person is an adult. In some cases, the person is a child. In other cases, the person is less than 12 years old. In some cases, the person is an elderly person. In other cases, the age of the person is 60 years or older. Other examples of subjects include laboratory animals such as mice, rats, dogs, cats, goats, sheep, pigs, and cattle. The experimental animal may be an animal model of a disorder, such as a transgenic mouse with hypertensive pathology.
By "pharmaceutically acceptable" is meant that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
The term "pharmaceutical composition" shall mean a composition comprising at least one active ingredient, whereby the composition is suitable for studying specific effective results in mammals (e.g. but not limited to humans). Those of ordinary skill in the art will understand and appreciate techniques suitable for determining whether an active ingredient has a desired effective result based on the needs of the skilled artisan.
As used herein, "therapeutically effective amount" or "effective amount" refers to the amount of an active compound or pharmaceutical agent that elicits the biological or medical response in a tissue, system, animal, subject, or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (1) prevention of disease; for example, preventing a disease, condition, or disorder in an individual who may be susceptible to the disease, condition, or disorder but who has not yet experienced or displayed the pathology or symptomatology of the disease, (2) inhibiting the disease; for example, inhibiting a disease, condition, or disorder in an individual experiencing or exhibiting the pathology or symptoms of the disease, condition, or disorder (i.e., arresting further development of the pathology and/or symptom), and (3) ameliorating the disease; for example, a disease, condition, or disorder (i.e., reversing pathology and/or symptoms) is ameliorated in an individual experiencing or exhibiting the pathology or symptoms of the disease, condition, or disorder.
The term "treatment" as used herein refers in some embodiments to therapeutic treatment, and in other embodiments to prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder, or disease, or to obtain a beneficial or desired clinical result. For purposes described herein, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; reducing the extent of a condition, disorder or disease; stabilization (i.e., not worsening) of the condition, disorder or disease state; delay onset of or slow progression of the condition, disorder or disease; improving a condition, disorder or disease state; and either detectable or undetectable remission (whether partial or complete) of a condition, disorder or disease, or enhancement or amelioration of a condition, disorder or disease. Treatment involves eliciting a clinically significant response without undue levels of side effects. Treatment also includes prolonging survival compared to the expected survival without treatment. The prophylactic benefit of treatment includes preventing the condition, slowing the progression of the condition, stabilizing the condition, or reducing the likelihood of the condition developing. As used herein, in some embodiments, "treating" includes prophylaxis.
The term "substantially identical" as used herein means that the powder X-ray diffraction pattern or differential scanning calorimetry pattern is different from those depicted herein, but falls within the limits of experimental error when considered by one of ordinary skill in the art.
Compound 1
Disclosed herein are (S) -N- (2-amino-1- (3-chloro-5-fluorophenyl) ethyl) -1- (5-methyl-2- ((tetrahydro-2H-pyran-4-yl) amino) pyrimidin-4-yl) -1H-imidazole-4-carboxamide:or a pharmaceutically acceptable salt thereof.
In some embodiments, the salt of compound 1 is a mandelate salt. In some embodiments, the salt of compound 1 is a benzenesulfonate salt. In some embodiments, the salt of compound 1 is the hydrochloride salt. In some embodiments, the salt of compound 1 is p-toluenesulfonate.
EGFR inhibitors
EGFR inhibitors are agents that bind to EGFR and slow or stop cell growth, and can be classified as Tyrosine Kinase Inhibitors (TKIs) or monoclonal antibodies.
TKIs are inhibitors that bind to tyrosine kinase domains in the epidermal growth factor receptor and stop EGFR activity. Examples include, but are not limited to, afatinib, dacatinib, erlotinib, gefitinib, lapatinib, lanitinib, lifatinib, mo Bo tinib, azatinib, lenatinib, octtinib, and vandetanib.
Monoclonal antibody inhibitors are agents that bind to the extracellular component of EGFR and prevent binding of epidermal growth factor to its own receptor, thereby preventing cell division. Examples include, but are not limited to, anti-angstrom Mo Tuoshan, cetuximab, clozotocet, nituzumab, and cetuximab.
In some embodiments, the EGFR inhibitor is afatinib, el Mo Tuoshan antibody, canetinib, cetuximab, dactyltinib, dapatinib, erlotinib, gefitinib, icotinib, lapatinib, lasatinib, lifenib, crizotinib, mo Bo tinib, natatinib, cetuximab, lenatinib, octyitinib, panitumumab, pelitinib, wave Ji Tini, tivozanib, rositinib, sapatinib, vandetanib, or valatinib.
In some embodiments, the EGFR inhibitor is AC480, AEE788, AG-1478, AG-18, AG-490, AST-1306, AV-412, AZ5104, AZD3759, BIBX 1382, CGP-52411, CL-387785, CNX-2006, CUDC-101, OSI-420, PD153035HCl, PD168393, TAK-285, tyrosine phosphorylation inhibitor (Tyrphostin) 9, tyrosine phosphorylation inhibitor AG 183, WHI-P154, WHI-P180, WZ3146, or WZ4002.
In some embodiments, the EGFR inhibitor is a small molecule inhibitor.
In some embodiments, the EGFR inhibitor is octreotide.
In some embodiments, the EGFR inhibitor is cetuximab.
In some embodiments, the EGFR inhibitor is afatinib.
In some embodiments, the EGFR inhibitor is dactinib.
In some embodiments, the EGFR inhibitor is erlotinib.
In some embodiments, the EGFR inhibitor is gefitinib.
In some embodiments, the EGFR inhibitor is lapatinib.
In some embodiments, the EGFR inhibitor is lanitinib.
In some embodiments, the EGFR inhibitor is lififensil.
In some embodiments, the EGFR inhibitor is Mo Bo tinib.
In some embodiments, the EGFR inhibitor is azatinib.
In some embodiments, the EGFR inhibitor is lenatinib.
In some embodiments, the EGFR inhibitor is vandetanib.
In some embodiments, the EGFR inhibitor is not an anti-EGFR antibody inhibitor.
Oritinib
OritinibAre small molecule EGFR tyrosine kinase inhibitors and are useful in the treatment of locally advanced or metastatic NSCLC. It was approved by the FDA at 11 months 2015 for specific treatment of metastatic NSCLC with EGFR exon 19 deletion or exon 21L858R mutation and EGFR T790M mutation positive NSCLC. Ornitinib is described by AstraZeneca as +. >And (5) selling. />
Cetuximab
Cetuximab is a chimeric monoclonal antibody EGFR inhibitor for use in the treatment of metastatic CRC and head and neck cancer. 7 months 2009, FDA approved cetuximab for the treatment of colon cancer with wild-type KRAS. Cetuximab is taken as Eli Lilly and CompanyAnd (5) selling.
BRAF inhibitors
BRAF inhibitors selectively target BRAF kinase, thereby interfering with MAPK signaling pathways that regulate melanoma cell proliferation and survival. BRAF inhibitors also have beneficial effects on the tumor microenvironment and anti-tumor immune response in BRAF mutant melanoma, thereby exerting an immunomodulatory effect on the MAPK pathway and promoting recognition of tumor cells by the immune system and enhancing anti-tumor T cell responses.
In some embodiments, the BRAF inhibitor is Kang Naifei ni.
In some embodiments, the BRAF inhibitor is dabrafenib.
Kang Naifei Ni
Kang Naifei NiIs a medicine for treating some melanoma. It is a small molecule BRAF inhibitor targeting a key enzyme in the MAPK signaling pathway. This pathway occurs in many different cancers, including melanoma and colorectal cancer. Month 6 of 2018, the FDA approved it in combination with bimetinib for the treatment of patients with unresectable or metastatic BRAF V600E or V600K mutation positive melanoma. Kang Naifei by Pfizer >And (5) selling.
Darafenib
DarafenibIs a drug for treating cancer associated with a mutated version of the gene BRAF. Darafenib acts as an inhibitor of the related enzyme B-Raf, which plays a role in regulating cell growth. Dabrafenib has clinical activity with a manageable safety profile in phase 1 and phase 2 clinical trials in patients with BRAF (V600) mutated metastatic melanoma. Darafenib is expressed by Novartis as +.>And (5) selling.
Combination of two or more kinds of materials
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof, and
(ii) EGFR inhibitors.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof, and
(ii) Ornitinib.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof, and
(ii) Cetuximab.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) An EGFR inhibitor; and
(iii) BRAF inhibitors.
In some embodiments, the EGFR inhibitor is a small molecule inhibitor.
In some embodiments, the EGFR inhibitor is afatinib, el Mo Tuoshan antibody, cetuximab, dactyltinib, erlotinib, gefitinib, lapatinib, lasatinib, lifenib, clomizotocet mab, mo Bo tinib, natatinib, cetuximab, lenatinib, octreotide, nimodib, or vandetanib. In some embodiments, the EGFR inhibitor is octreotide. In some embodiments, the EGFR inhibitor is afatinib. In some embodiments, the EGFR inhibitor is dactinib. In some embodiments, the EGFR inhibitor is erlotinib. In some embodiments, the EGFR inhibitor is gefitinib. In some embodiments, the EGFR inhibitor is lapatinib. In some embodiments, the EGFR inhibitor is lanitinib. In some embodiments, the EGFR inhibitor is lififensil. In some embodiments, the EGFR inhibitor is Mo Bo tinib. In some embodiments, the EGFR inhibitor is azatinib. In some embodiments, the EGFR inhibitor is lenatinib. In some embodiments, the EGFR inhibitor is vandetanib. In some embodiments, the EGFR inhibitor is cetuximab. In some embodiments, the EGFR inhibitor is not an anti-EGFR antibody inhibitor. In some embodiments, the BRAF inhibitor is dabrafenib, kang Naifei ni, regafinib, sorafenib, or vemurafenib.
In some embodiments, the BRAF inhibitor is dabrafenib. In some embodiments, the BRAF inhibitor is Kang Naifei ni.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Cetuximab; and
(iii) Kang Naifei Ni.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of (i) compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Cetuximab; and
(iii) Darafenib.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Ornitinib; and
(iii) Kang Naifei Ni.
Disclosed herein is a method of treating cancer in a subject in need thereof, the method comprising: administering to a subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Ornitinib; and
(iii) Darafenib.
Further combination of
In some embodiments, the method comprises administering an additional MAPK pathway inhibitor. Without being bound by theory, inhibition of MAPK signaling in cancer cells may lead to down-regulation of PD-L1 expression and increase the likelihood that cancer cells will be detected by the immune system. Such a third MAPK pathway inhibitor may be based on other mutations in the protein in the MAPK pathway. In some embodiments, the additional MAPK pathway inhibitor inhibits a protein in the MAPK pathway. In some embodiments, the additional MAPK pathway inhibitor inhibits a protein other than the MAPK pathway. In some embodiments, the additional MAPK pathway inhibitor is a KRAS inhibitor, NRAS inhibitor, HRAS inhibitor, PDGFRA inhibitor, PDGFRB inhibitor, MET inhibitor, FGFR inhibitor, ALK inhibitor, ROS1 inhibitor, TRKA inhibitor, TRKB inhibitor, TRKC inhibitor, EGFR inhibitor, IGFR1R inhibitor, GRB2 inhibitor, SOS inhibitor, ARAF inhibitor, BRAF inhibitor, RAF1 inhibitor, MEK2 inhibitor, c-Mycv, CDK4/6, inhibitor CDK2 inhibitor, FLT3 inhibitor, or ERK1/2 inhibitor. Exemplary MAPK pathway inhibitors include, but are not limited to, adaglazeb, afatinib, ASTX029, bimatinib, cobitinib, dacatinib, erlotinib, gefitinib, ji Ruiti, lapatinib, LTT462, LY3214996, cetuximab, lenatinib, nimotuzumab, pamazetinib, sematinib, sotolazeb, trimatinib, ulitinib, vandetanib, and valfefilib.
In some embodiments, the additional MAPK pathway inhibitor is adaglazeb. In some embodiments, the additional MAPK pathway inhibitor is afatinib. In some embodiments, the additional MAPK pathway inhibitor is bimetanib. In some embodiments, the additional MAPK pathway inhibitor is cobicitinib. In some embodiments, the additional MAPK pathway inhibitor is dactinib. In some embodiments, the additional MAPK pathway inhibitor is erlotinib. In some embodiments, the additional MAPK pathway inhibitor is gefitinib. In some embodiments, the additional MAPK pathway inhibitor is gefitinib. In some embodiments, the additional MAPK pathway inhibitor is lapatinib. In some embodiments, the additional MAPK pathway inhibitor is LTT462. In some embodiments, the additional MAPK pathway inhibitor is LY3214996. In some embodiments, the additional MAPK pathway inhibitor is rituximab. In some embodiments, the additional MAPK pathway inhibitor is lenatinib. In some embodiments, the additional MAPK pathway inhibitor is nituzumab. In some embodiments, the additional MAPK pathway inhibitor is palbociclib. In some embodiments, the additional MAPK pathway inhibitor is semantenib. In some embodiments, the additional MAPK pathway inhibitor is sotorubin. In some embodiments, the additional MAPK pathway inhibitor is trametinib. In some embodiments, the additional MAPK pathway inhibitor is ulitinib. In some embodiments, the additional MAPK pathway inhibitor is vandetanib.
Cancer of the human body
Disclosed herein are methods of treating cancer using the combinations disclosed herein.
"cancer" refers to all types of cancers, neoplasms, or malignant tumors found in mammals (e.g., humans), including, but not limited to, leukemia, lymphoma, myeloma, carcinoma, and sarcoma. Exemplary cancers that may be treated with the compounds or methods provided herein include brain cancer, glioma, glioblastoma, neuroblastoma, prostate cancer, colorectal cancer, pancreatic cancer (such as pancreatic adenocarcinoma, PDAC), medulloblastoma, melanoma, cervical cancer, gastric cancer, ovarian cancer, lung cancer, head cancer, hodgkin's disease, and non-hodgkin's lymphoma. Exemplary cancers that may be treated with the compounds or methods provided herein include blood cancer, thyroid cancer, cancer of the endocrine system, brain cancer, breast cancer, cervical cancer, colon cancer, head and neck cancer, liver cancer, kidney cancer, lung cancer, ovarian cancer, pancreatic cancer, rectal cancer, stomach cancer, and uterine cancer. Further examples include thyroid cancer, cholangiocarcinoma, pancreatic adenocarcinoma, skin melanoma, colon adenocarcinoma, rectal adenocarcinoma, gastric adenocarcinoma, esophageal cancer, head and neck squamous cell carcinoma, breast invasive carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, non-small cell lung cancer, mesothelioma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, ovarian cancer, rhabdomyosarcoma, primary thrombocythemia, primary macroglobulinemia, primary brain tumor, malignant islet tumor (malignant pancreatic insulanoma), malignant carcinoid, bladder cancer, premalignant skin lesions, testicular cancer, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenocortical carcinoma, endocrine or exocrine pancreatic tumor, medullary thyroid cancer (medullary thyroid cancer), medullary thyroid cancer (medullary thyroid carcinoma), melanoma, colorectal cancer, papillary thyroid cancer, hepatocellular carcinoma or prostate cancer.
In some embodiments, the cancer has a class 1B-Raf mutation.
In some embodiments, the cancer has at least one of EGFR, KRAS, BRAF (e.g., BRAF class III) and/or NF 1 (e.g., loss of function) mutation.
In some embodiments, the mutant B-Raf comprises a V600 mutation. In some embodiments, the mutant of B-Raf comprises the mutation V600E. In some embodiments, the mutation is V600K. In some embodiments, the mutation is V600D. In some embodiments, the mutation is V600L. In some embodiments, the mutation is V600R. In some embodiments, the cancer is a BRAF V600E or V600K mutant tumor.
In some embodiments, the cancer is a mitogen-activated protein kinase (MAPK) pathway-driven cancer.
In some embodiments, the cancer is a BRAF-driven cancer, HRAS-driven cancer, or NRAS-driven cancer.
In some embodiments, the cancer comprises at least one cancer cell driven by deregulated ERK.
In some embodiments, the cancer has at least one mutation in the RAS. In some embodiments, the cancer has at least one mutation in RAF. In some embodiments, the cancer has at least one mutation in MEK.
In some embodiments, the cancer has a G12C KRAS mutation. In some embodiments, the cancer has a G12D KRAS mutation. In some embodiments, the cancer has a G12S KRAS mutation. In some embodiments, the cancer has a G12V KRAS mutation. In some embodiments, the cancer has a G13D KRAS mutation. In some embodiments, the cancer has a Q16H KRAS mutation. In some embodiments, the cancer has a Q16KKRAS mutation. In some embodiments, the cancer has a Q61R NRAS mutation.
In some embodiments, the cancer is a pan-carcinoma that does not receive MAPKm/MAPKi.
In some embodiments, the cancer comprises one or more EGFR mutations selected from the group consisting of: increased EGFR gene copy, EGFR gene amplification, chromosome 7 multimerity, L858R, exon 19 deletion/insertion, L718Q, L861 719A, G724S, V765A, T783A, exon 20 insertion, EGFR splice variants (Viii, vvi and Vii), A289D, A289T, A289V, G598 458 598V, T790M, C797 6279S and S768I. In some embodiments, the cancer comprises one or more EGFR mutations selected from the group consisting of L858R, exon 19 deletion, and T790M.
In some embodiments, the cancer is a solid tumor. In some embodiments, the solid tumor is an advanced or metastatic solid tumor.
In some embodiments, the cancer is non-small cell lung cancer (NSCLC), melanoma, pancreatic cancer, salivary gland tumor, thyroid cancer, colorectal cancer (CRC), or esophageal cancer.
In some embodiments, the cancer is colorectal cancer (CRC), pancreatic Ductal Adenocarcinoma (PDAC), cholangiocarcinoma, appendiceal carcinoma, gastric cancer, esophageal cancer, non-small cell lung cancer (NSCLC), head and neck cancer, ovarian cancer, uterine cancer, acute Myelogenous Leukemia (AML), or melanoma.
In some embodiments, the cancer is gastrointestinal cancer. In some embodiments, the gastrointestinal cancer is anal cancer, cholangiocarcinoma, colon cancer, rectal cancer, esophageal cancer, gallbladder cancer, liver cancer, pancreatic cancer, small intestine cancer, or stomach cancer.
In some embodiments, the cancer is non-small cell lung cancer (NSCLC). In some embodiments, the NSCLC is an EGFR mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12C mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12D mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12S mutant NSCLC. In some embodiments, the NSCLC is a KRAS G12V mutant NSCLC. In some embodiments, the NSCLC is a KRAS G13D mutant NSCLC. In some embodiments, the NSCLC is a KRAS Q61H mutant NSCLC. In some embodiments, the NSCLC is a KRAS Q61K mutant NSCLC.
In some embodiments, the NSCLC is a RAS Q61R mutant NSCLC. In some embodiments, the cancer is NSCLC that does not receive MAPKm/MAPKi. In some embodiments, the cancer is BRAFi treated V600 NSCLC. In some embodiments, the cancer is KRAS treated G12C NSCLC. In some embodiments, the cancer is KRAS treated G12D NSCLC. In some embodiments, the cancer is KRAS treated G12S NSCLC. In some embodiments, the cancer is KRAS treated G12V NSCLC. In some embodiments, the cancer is KRAS treated G13DNSCLC. In some embodiments, the cancer is KRAS treated Q61H NSCLC. In some embodiments, the cancer is KRAS treated Q61K NSCLC. In some embodiments, the cancer is NRAS treated Q61R NSCLC.
In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is pancreatic cancer that does not receive MAPKm/MAPKi. In some embodiments, the cancer is Pancreatic Ductal Adenocarcinoma (PDAC). In some embodiments, PDAC is indicated by a KRAS G12V mutation.
In some embodiments, the cancer is melanoma. In some embodiments, the melanoma is a BRAF V600E or V600K mutant tumor. In some embodiments, the cancer is BRAFi treated V600 melanoma.
In some embodiments, the cancer is a salivary gland tumor.
In some embodiments, the cancer is thyroid cancer.
In some embodiments, the cancer is colorectal cancer (CRC). In some embodiments, the CRC is a BRAF V600E CRC. In some embodiments, the CRC is a KRAS mutant CRC.
In some embodiments, the CRC is a KRAS G12C mutant CRC. In some embodiments, the CRC is a KRAS G12D mutant CRC. In some embodiments, the CRC is a KRAS G12S mutant CRC. In some embodiments, the CRC is a KRAS G12V mutant CRC. In some embodiments, the CRC is a KRAS G13D mutant CRC. In some embodiments, the CRC is a KRAS Q61H mutant CRC. In some embodiments, the CRC is a KRAS Q61K mutant CRC. In some embodiments, the CRC is an NRAS mutant CRC. In some embodiments, the CRC is an NRAS Q61R mutant CRC.
In some embodiments, the cancer is esophageal cancer.
In some embodiments, the cancer has one or more acquired mutations. In some embodiments, the acquired mutation results from a first line therapy. In some embodiments, the first line treatment is an EGFR inhibitor. In some embodiments, the EGFR inhibitor is octreotide. In some embodiments, the EGFR inhibitor is cetuximab. In some embodiments, the cancer is a solid tumor cancer. In some embodiments, the cancer is NSCLC.
In some embodiments, the acquired mutation is an acquired EGFR mutation. In some embodiments, the acquired EGFR mutation is C797X. In some embodiments, the acquired EGFR mutation is L718Q. In some embodiments, the acquired EGFR mutation is EGFR amplification. In some embodiments, the acquired EGFR mutation is G724S. In some embodiments, the acquired mutation is S768I.
In some embodiments, the acquired mutation is an acquired amplification mutation. In some embodiments, the acquired mutation is MET gene amplification. In some embodiments, the acquired mutation is HER2 gene amplification.
In some embodiments, the acquired mutation is an acquired oncogenic fusion. In some embodiments, the acquired oncogenic fusion is SPTBN1-ALK. In some embodiments, the acquired oncogenic fusion is a RET fusion. In some embodiments, the acquired oncogenic fusion is a BRAF fusion.
In some embodiments, the acquired mutation is an acquired MAPK-PI3K mutation. In some embodiments, the acquired MAPK-PI3K mutation is BRAF-V600E. In some embodiments, the acquired MAPK-PI3K mutation is PI3KCA. In some embodiments, the acquired MAPK-PI3K mutation is KRAS. In some embodiments, the acquired MAPK-PI3K mutation is HER2.
Administration of drugs
In one aspect, the compositions described herein are used to treat diseases and conditions described herein. Furthermore, a method for treating any disease or condition described herein in a subject in need of such treatment comprises administering to the subject a therapeutically effective amount of the composition.
The dosage of the compositions described herein may be determined by any suitable method. The Maximum Tolerated Dose (MTD) and the Maximum Response Dose (MRD) of compound 1 or a pharmaceutically acceptable salt thereof can be determined by established animal and human protocols and examples described herein. For example, toxicity and therapeutic efficacy of compound 1 or a pharmaceutically acceptable salt thereof can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective to 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio between LD50 and ED50. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds is preferably within a circulating concentration range, including the ED50 with minimal toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration employed. Additional relative doses expressed as percentages of the maximum response dose or maximum tolerated dose are readily obtained via the protocol.
In some embodiments, the amount of a given formulation comprising compound 1 or a pharmaceutically acceptable salt thereof corresponding to such amount varies depending on factors such as the molecular weight of the particular salt or form, the disease condition and severity thereof, the identity (e.g., age, weight, sex) of the subject or host in need of treatment, and the like, but may still be determined based on the particular circumstances surrounding the case, including, for example, the particular agent administered, the type of liquid formulation, the condition being treated, and the subject or host being treated.
In some embodiments, kang Naifei Ni is administered in an amount of about 100 mg/day to 500 mg/day. In some embodiments, kang Naifei Ni is administered in an amount of about 450 mg/day. In some embodiments, kang Naifei Ni is administered in an amount of about 300 mg/day. In some embodiments, kang Naifei Ni is administered in an amount of about 250 mg/day. In some embodiments, kang Naifei Ni is administered in an amount of about 225 mg/day. In some embodiments, kang Naifei Ni is administered in an amount of about 200 mg/day. In some embodiments, kang Naifei Ni is administered in an amount of about 150 mg/day.
In some embodiments, cetuximab is administered at 400mg/m once a week 2 Applied over 120 minutes, then at 250mg/m 2 Applied over a period of 60 minutes. In some embodiments, cetuximab is administered at 500mg/m once every two weeks 2 And (3) application. In some embodiments, cetuximab is administered at 400mg/m once every two weeks 2 And (3) application. In some embodiments, cetuximab is administered at 300mg/m once every two weeks 2 And (3) application. In some embodiments, cetuximab is administered at 200mg/m once every two weeks 2 And (3) application.
In some embodiments, the amount of compound 1, or a pharmaceutically acceptable salt thereof, as described herein is relative to the free base equivalent of compound 1.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered orally.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day to about 300 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of 25 mg/day to 150 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day, about 50 mg/day, about 75 mg/day, about 100 mg/day, about 125 mg/day, about 150 mg/day, about 175 mg/day, about 200 mg/day, about 225 mg/day, or about 250 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day, about 50 mg/day, about 100 mg/day, or about 150 mg/day.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 200mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 100mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 50mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 50mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 50mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 50mg to about 200mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 50mg to about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 50mg to about 100mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg to about 200mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg to about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 150mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 150mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 150mg to about 200mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 175mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 175mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 175mg to about 200mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 200mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 200mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 225mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 225mg to about 250mg twice daily, one day per week (BID-QW). In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 300mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 50mg to about 250mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg to about 300mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg to about 250mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 150mg to about 300mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 150mg to about 250mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 100mg, once per week (QW). In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 150mg, once per week (QW). In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 200mg, once per week (QW). In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 250mg, once per week (QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 300mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 250mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 50mg, about 75mg, about 100mg, about 125mg, about 150mg, about 175mg, about 200mg, about 225mg, or about 250mg, twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 50mg, about 100mg, about 125mg, or about 150mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 125mg twice daily, one day per week (BID-QW).
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 250mg, once a day, one day a week.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 30mg, 40mg, 50mg, about 60mg, about 70mg, about 75mg, about 80mg, about 85mg, about 90mg, about 95mg, about 100mg, about 105mg, about 110mg, about 115mg, about 120mg, about 125mg, about 130mg, about 140mg, about 150mg, about 160mg, about 170mg, about 175mg, about 180mg, about 190mg, about 200mg, about 210mg, about 220mg, about 225mg, about 230mg, about 240mg, about 250mg, about 260mg, about 270mg, about 280mg, about 290mg, or about 300 mg.
In some embodiments, each of the above amounts may be administered as QD, QW, BID, BID-QD or BID-QW.
Application of
Compound 1 or a pharmaceutically acceptable salt thereof and a combination partner (partner) described herein are administered at the dosages described herein or at other dosage levels and compositions determined and considered by the medical practitioner. In certain embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered for prophylactic and/or therapeutic treatment. In certain therapeutic applications, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a patient already suffering from a disease in an amount sufficient to cure the disease or at least partially prevent or ameliorate symptoms. The amount effective for this use will depend on the age of the patient, the severity of the disease, the previous treatment, the health of the patient, the weight and response to the composition, and the judgment of the treating physician. The therapeutically effective amount is optionally determined by methods including, but not limited to, up-dosing clinical trials.
In prophylactic applications, the compositions described herein are administered to a patient susceptible to or otherwise at risk of a particular disease (e.g., cancer). Such an amount is defined as a "prophylactically effective amount or dose". In this use, the precise amount will also depend on the age, health, weight, etc. of the patient. When used in a patient, the effective amount for such use will depend on the risk or susceptibility to developing the particular disease, the previous treatment, the patient's health and response to the composition, and the discretion of the treating physician.
In certain embodiments, wherein the condition of the patient is not improved, the compositions described herein are administered chronically (i.e., for an extended period of time, including throughout the life of the patient) at the discretion of the physician, in order to ameliorate or otherwise control or limit the symptoms of the patient's disease. In other embodiments, the administration of the composition is continued until a complete or partial response to the disease.
In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, and a combination partner described herein, are administered once daily. In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, and a combination partner described herein, are administered twice daily. In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, and a combination partner described herein, are administered three times daily.
In some embodiments, kang Naifei ni is administered once daily. In some embodiments, kang Naifei ni is administered twice daily. In some embodiments, kang Naifei ni is administered three times per day.
In some embodiments, cetuximab is administered once every three weeks. In some embodiments, cetuximab is administered once every four weeks. In some embodiments, cetuximab is administered once every five weeks. In some embodiments, cetuximab is administered once every six weeks.
In some embodiments, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a subject in a fasted state. Fasted state refers to a subject that is not fed or fasted for a period of time. Typical fasted periods include no feeding for at least 4 hours, at least 6 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, and at least 16 hours. In some embodiments, compound 1, or a pharmaceutically acceptable salt thereof, is administered to a subject in a fasted state for at least 8 hours. In other embodiments, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a subject in a fasted state for at least 10 hours. In still other embodiments, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a subject in a fasted state for at least 12 hours. In other embodiments, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a subject who has fasted overnight.
In other embodiments, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a subject in a fed state. Fed state refers to a subject that has been fed or has been fed. In certain embodiments, the composition is administered to a subject in a fed state at 5 minutes post-meal, 10 minutes post-meal, 15 minutes post-meal, 20 minutes post-meal, 30 minutes post-meal, 40 minutes post-meal, 50 minutes post-meal, 1 hour post-meal, or 2 hours post-meal. In certain instances, compound 1 or a pharmaceutically acceptable salt thereof is administered to a subject in a fed state 30 minutes after a meal. In other cases, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered to a subject in a fed state 1 hour after a meal. In a further embodiment, compound 1 or a pharmaceutically acceptable salt thereof is administered to a subject in combination with food.
The length of the treatment period depends on the treatment administered. In some embodiments, the length of the treatment period ranges from 2 to 6 weeks. In some embodiments, the length of the treatment period ranges from 3 to 6 weeks. In some embodiments, the length of the treatment period ranges from 3 to 4 weeks. In some embodiments, the length of the treatment period is 3 weeks (or 21 days). In some embodiments, the length of the treatment period is 4 weeks (28 days). In some embodiments, the length of the treatment period is 5 weeks (35 days). In some embodiments, the length of the treatment period is 56 days. In some embodiments, the treatment period lasts for 1, 2, 3, 4, or 5 weeks. In some embodiments, the treatment period lasts for 3 weeks. In some embodiments, the treatment period lasts for 4 weeks. In some embodiments, the treatment period lasts for 5 weeks. The number of therapeutic doses scheduled in each cycle also varies depending on the drug administered.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered at a 28 day period. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in a plurality of 28-day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least one 28-day cycle. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least two 28-day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least three 28-day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least four 28-day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least five 28-day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least six 28-day cycles.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-7 of each 28-day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-14 of each 28-day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-21 of each 28-day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-28 of each 28-day cycle.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 1 of a 28 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 8 of the 28 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 15 of a 28 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 22 of the 28 day cycle. In some embodiments of the method of treating cancer, compound 1, or a pharmaceutically acceptable salt thereof, is not administered twice a day on day 22 of the 28 day cycle.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice daily on days 1, 8 and 15 of a 28 day cycle.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is not administered on days 2-7, 9-14, 16-21, 23-28 of the 28 day cycle.
In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in a 35 day period. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in a plurality of 35 day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered at least one 35 day period. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least two 35 day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least three 35 day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least four 35 day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least five 35 day cycles. In some embodiments of the methods of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof and a combination partner described herein are administered in at least six 35 day cycles.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-7 of each 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-14 of each 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-21 of each 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-28 of each 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1-35 of each 35 day cycle.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 1 of a 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 8 of the 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 15 of a 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 22 of the 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice a day on day 29 of the 35 day cycle. In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is not administered twice a day on day 29 of the 35 day cycle.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is administered twice daily on days 1, 8, 15 and 22 of a 35 day cycle.
In some embodiments of the method of treating cancer, compound 1 or a pharmaceutically acceptable salt thereof is not administered on days 2-7, 9-14, 16-21, 23-28, and 30-35 of the 28 day cycle.
Patient selection
In some embodiments of the method of treating cancer, the subject's age is 18 years. In another embodiment, the subject is 18 years of age or older. In another embodiment, the subject is no more than 99 years old.
In some embodiments of the method of treating cancer, the subject is willing and able to give written informed consent.
In some embodiments of the method of treating cancer, the subject has a histologically or cytologically confirmed metastatic CRC carrying the applicable mutation based on an analytically validated assay performed on tumor tissue in a certified test laboratory. In some embodiments, the subject has a BRAFm V600E mutation. In some embodiments, the subject has a KRAS mutation. In some embodiments, the subject has NRAS mutations.
In some embodiments of the method of treating cancer, the subject has an advanced NSCLC bearing one or more EGFR mutations that is histologically or cytologically confirmed.
In some embodiments of the method of treating cancer, the subject has sufficient bone marrow and organ function.
In some embodiments of the method of treating cancer, the subject's eastern tumor cooperative group (Eastern Cooperative Oncology Group, ECOG) exhibits a status of 0 or 1.
In some embodiments of the method of treating cancer, the subject is willing to adhere to all of the regimen required access, assessment and procedures.
In some embodiments of the method of treating cancer, the subject is able to swallow oral medications.
In some embodiments of the method of treating cancer, the subject has not received prior treatment with an inhibitor of RAS, MEK, or ERK.
In some embodiments of the method of treating cancer, the subject does not receive concurrent treatment with any systemic anti-cancer therapy for NSCLC.
In some embodiments of the method of treating cancer, the subject has not received prior Cancer Immunotherapy (CIT) unless prior to study recruitment, a non-CIT containing regimen follows CIT.
In some embodiments of the method of treating cancer, the subject does not receive anti-cancer treatment within 21 days of recruitment, except for octenib, which may continue during the screening period.
In some embodiments of the method of treating cancer, the subject has no unacceptable history of toxicity for the treatment of octenib.
In some embodiments of the method of treating cancer, the subject does not receive an anti-cancer therapy no more than 21 days prior to the first dose of the study drug or combination of drugs.
In some embodiments of the method of treating cancer, the subject does not receive an anti-cancer therapy 4 half-lives before the first dose of the study drug or drug combination.
In some embodiments of the method of treating cancer, the subject does not receive palliative radiation for less than or equal to 7 days prior to the first dose of the study drug or combination of drugs.
In some embodiments of the method of treating cancer, the subject has not had or has not been diagnosed with symptomatic brain metastasis or a pia mater disease.
In some embodiments of the method of treating cancer, the subject is not suffering from or diagnosed with a gastrointestinal condition that may affect the absorption of oral drugs.
In some embodiments of the method of treating cancer, the subject does not have or is not diagnosed with an active infection, or a history of HIV infection, hepatitis b virus, or hepatitis c virus, in need of systemic treatment.
In some embodiments of the method of treating cancer, the subject has no history of chronic inflammatory bowel disease or Crohn's disease requiring medical intervention (e.g., immunomodulation, immunosuppressant drugs or surgery) no more than 12 months prior to the first dose of study drug or drug combination.
In some embodiments of the method of treating cancer, the subject has no active and clinically significant interstitial lung disease or pneumonia.
In some embodiments of the method of treating cancer, the subject is free of impaired cardiovascular function or clinically significant cardiovascular disease.
In some embodiments of the method of treating cancer, the subject has no history of thromboembolism or cerebrovascular events less than or equal to 6 months prior to the first dose of study drug or combination of drugs.
In some embodiments of the method of treating cancer, the subject is free of a history or current evidence of Retinal Pigment Epithelial Detachment (RPED), central serous retinopathy, retinal Vein Occlusion (RVO), or RPED or RVO-inducing factors.
In some embodiments of the method of treating cancer, the subject does not undergo major surgery within 28 days of study recruitment or is expected to undergo no major surgery during the study treatment.
In some embodiments of the method of treating cancer, the subject is not known intolerance or contraindication to Kang Naifei ni, cetuximab, or palbociclib.
In some embodiments of the method of treating cancer, the subject has no known intolerance or contraindication to octenib.
In some embodiments of the method of treating cancer, the subject is not pregnant or is not lactating.
In some embodiments of the method of treating cancer, the subject does not have any evidence of severe or uncontrolled systemic disease or any other significant clinical condition or laboratory findings that render the patient unsuitable for participation in the study.
Examples
Example 1: in vivo assay (Compound 1+ Kang Naifei Ni/cetuximab)
Vehicle/control article
Vehicle/control preparations were prepared, 0.5% methylcellulose and 0.1% tween 80 or 100mM acetic acid in deionized water, pH adjusted to 4.8-5.0, and stored at ambient conditions throughout the study period.
Configuration of test article
Test article compound 1 was freshly prepared weekly in a vehicle of 0.5% methylcellulose and 0.1% tween 80 and stored under ambient conditions. The combination agent Kang Naifei Ni was freshly prepared weekly in 0.5% methylcellulose and 0.5% Tween 80 vehicle and stored at 2-8deg.C. The combination agent cetuximab was diluted with 1x PBS and stored at 2-8 ℃ prior to dosing.
Animals
Female Balb/c nude mice were purchased from beijing velarihua laboratory animal technologies limited (Beijing Vital River Laboratory Animal Technology co., ltd.). Mice were housed in an animal feeding facility in an environment free of Specific Pathogens (SPF) and allowed to adapt to their new environment for at least 3 days before any experiments were initiated. Mice were 6-8 weeks of age at implantation.
All procedures related to animal handling, care and treatment in this study were performed according to protocols and guidelines approved by the institutional animal care and use committee (Institutional Animal Care and Use Committee, IACUC) of GenenDesign and WuXi AppTec. Animal facilities and procedures operate under the standards of the laboratory animal care and use guidelines (Guide for the Care and Use of Laboratory Animals) (NRC, 2011) and are certified by the laboratory animal care assessment and certification association (Association for Assessment and Accreditation of Laboratory Animal Care, AAALAC). Specifically, all parts of the study conducted in GenenDesign and WuXi AppTec followed the study protocol reviewed and approved by IACUC and the Standard Operating Program (SOP) applicable.
Preparation of xenograft models
RKO is a portable BRAF V600E Mutant human CRC tumor cell lines. RKO cell lines were purchased from American type culture Collection (American Type Culture Collection)/(America)CRL-2577 TM ). RKO cells were cultured in a medium containing EMEM plus 10% Fetal Bovine Serum (FBS) supplemented with nonessential amino acids at 37℃under a culture atmosphere containing 5% CO 2 Is a gas in the air chamber. Will contain 2X 10 mixed with 50% matrigel 6 RKO cells in 200. Mu.L cell suspension of individual cells were subcutaneously implanted into mice. When the tumor volume reached an average of 200mm 3 At this time, tumor-bearing mice were randomized into different groups of 8 mice each and treatment was started on the day of randomization.
WiDr is a portable BRAF V600E Mutant human CRC tumor cell lines. The WiDr cell line was purchased from european collection of certified cell cultures (European Collection of Authenticated Cell Cultures) (ECACC, 85111501). WiDr cells were cultured in a medium containing EMEM (EBSS) plus 10% Fetal Bovine Serum (FBS), 2mM glutamine and supplemented with 1% non-essential amino acid (NEAA) at 37℃under a culture atmosphere containing 5% CO 2 Is a gas in the air chamber. Will contain 5X 10 mixed with 50% matrigel 6 WiDr cells in 200. Mu.L cell suspension of individual cells were subcutaneously implanted into mice. When the tumor volume reached an average of 190mm 3 At this time, tumor-bearing mice were randomized into different groups of 8 mice each and treatment was started on the day of randomization.
Treatment of
In the monotherapy treatment group, mice were dosed by oral administration of vehicle control solution, compound 1, kang Naifei ni or cetuximab. Mice were dosed by oral administration of a combination comprising compounds 1 and Kang Naifei ni and compound 1 and Kang Naifei nixib. The dosing volume of each compound was 5mL/kg and the BID protocol was 8 hours apart. In the combination of compound 1 and Kang Naifei, compound 1 was administered 1 hour after Kang Naifei. In the combination of compound 1 and Kang Naifei nigaxib, compound 1 was administered 1 hour after Kang Naifei nig and cetuximab was administered 1 hour after compound 1QD or the first dose BID. In addition to the conventional food and water supply, at least two mice therein showed >Dietgel (ClearH 2O, US) was added to the 10% BWL cage. Tumor volume reached 2,000mm at the end of 4 weeks of treatment or in vehicle control group 3 The study was terminated at that time.
Results
In BRAF V600E CRC CDX model RKO, compound 1 exhibited 82% Tumor Growth Inhibition (TGI) as monotherapy (p-value < 0.001), 88% TGI in combination with Kang Naifei ni (p-value < 0.001), and 93% TGI in combination with Kang Naifei ni and cetuximab (p-value < 0.001), as shown in the tumor growth curve of fig. 1A.
In BRAF V600E CRC CDX model WiDr, compound 1 exhibited 102% TGI as monotherapy (p-value < 0.001), 109% TGI in combination with Kang Naifei ni (p-value < 0.001), and 111% TGI in combination with Kang Naifei ni and cetuximab (p-value < 0.001), as shown in the tumor growth curve of fig. 1B.
Compound 1+ Kang Naifei ni and compound 1+ Kang Naifei ni/cetuximab refractory BRAF at Kang Naifei ni/cetuximab (EC) V600E CDX models exhibit in vivo combinatorial benefits.
Example 2: in vivo assay (Compound 1+Ornitinib)
Vehicle/control article
Vehicle/control preparations were prepared, 0.5% methylcellulose and 0.1% tween 80 or 100mM acetic acid in deionized water, pH adjusted to 4.8-5.0, and stored at ambient conditions throughout the study period.
Configuration of test article
Test article compound 1 was freshly prepared weekly in a vehicle of 0.5% methylcellulose and 0.1% tween 80 and stored under ambient conditions. The combination agent, octenib, was prepared weekly in a vehicle of 0.5% HPMC and 0.1% tween 80 and stored under ambient conditions.
Animals
Female Balb/c nude mice were purchased from Beijing Vietnam Lihua laboratory animal technologies Co. Mice were housed in an environment free of Specific Pathogens (SPF) in an animal farm facility and allowed to adapt to their new environment for at least 3 days before any experiments were initiated. Mice were 6-8 weeks of age at implantation.
All procedures related to animal handling, care and treatment in this study were performed according to protocols and guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of GenenDesign and WuXi AppTec. Animal facilities and procedures operate under the standards of the laboratory animal care and use guidelines (NRC, 2011) and are certified by the laboratory animal care assessment and certification association (AAALAC). Specifically, all parts of the study conducted in GenenDesign and WuXi AppTec followed the study protocol reviewed and approved by IACUC and the Standard Operating Program (SOP) applicable.
Preparation of xenograft models
LUN2355-214 PDX was established at GenenDesign for preclinical efficacy studies. The PDX model was derived from a 49 year old male chinese NSCLC patient. EGFR L858R, T790 and C797 mutations in the LUN2355-214 PDX model were confirmed by whole exome sequencing and PCR sequencing. LUN2355-128-33PDX was established at GenenDesign for preclinical efficacy studies. The PDX model was derived from a 49 year old male chinese NSCLC patient. EGFR L858R and T790 mutations in the LUN2355-128-33PDX model were confirmed by whole-exome sequencing and PCR sequencing. Both LUN2355-214 and LUN2355-128-33PDX models were treated and derivatized after long-term treatment with octenib and then used in combination efficacy studies of compound 1 with combination agents. PDX tumor fragments (15-30 mm) 3 ) The mice were implanted subcutaneously. When the tumor volume reached an average of 200mm 3 At this time, tumor-bearing mice were randomized into different groups of 8 mice each and treatment was started on the day of randomization.
Treatment of
In the monotherapy treatment group, mice are dosed by oral administration of a vehicle control solution, compound 1 or octreotide. Mice were dosed by oral administration comprising a combination of compound 1 and octreotide. The dosing volume of each compound was 5mL/kg and the BID protocol was 8 hours apart. In the combination of compound 1 and octreotide, compound 1 was administered 1 hour after the administration of octreotide. In addition to the conventional food and water supply, at least two mice therein showed >Dietgel (ClearH 2O, US) was added to the 10% BWL cage. Tumor volume reached 2,000mm at the end of 4 weeks of treatment or in vehicle control group 3 The study was terminated at that time.
Results
Compound 1 and octenib showed in vivo combinatorial benefits in the octenib refractory EGFR mutant PDX model (see, e.g., tumor growth curves for LUN 2355-214 (fig. 2A) and LUN 2355-128-33 (fig. 2B) PDX models).
Example 3: 1b/2 phase study of Compound 1, cetuximab and Kang Naifei Ni targeting the MAPK pathway in patients with advanced gastrointestinal malignancies
Study design
This phase 1b/2 sub-study will evaluate the safety, clinical pharmacology, and primary efficacy of QW administered compound 1 in combination with Kang Naifei ni and cetuximab in patients with previously treated BRAF V600E CRC. Part 1 will determine the recommended weekly (QW) dose (RD) of compound 1 in combination with once daily (QD) Kang Naifei ni and once every two weeks (Q2W) cetuximab. Part 2 will further evaluate RD from part 1 in a larger patient group. If open, part 3 will determine the RD of Compound 1QD combined with Kang Naifei NiQD and cetuximab Q2W. Part 4 will further evaluate RD from part 3 in a larger patient group. The decision to initiate part 3 will depend on the examination of the data from part 1. Only 1 RD from part 1 or part 3 will be selected for expansion in part 2 or part 4, respectively.
Up to about 296 patients with BRAF V600E CRC can be enrolled in this sub-study. In parts 1 and 3, up to about 36 patients with evaluable safety can be enrolled in each dose escalation part. Patients who were withdrawn from the study during the 28-day Dose Limiting Toxicity (DLT) evaluation period for reasons unrelated to treatment-induced toxicity will be replaced. In part 2 or part 4 (only one of these parts will be selected for expansion), up to about 200 efficacy evaluable patients (receiving at least 1 dose of compound 1 and having at least 1 post-dose tumor assessment) can be enrolled in the applicable part, with up to about 100 efficacy evaluable patients in each group. Assuming that about 10% of the dosed patients were withdrawn from the study prior to tumor assessment after the first dose, the dose-extending group could recruit and dose up to about 224 patients.
Parts 1 and 3 (dose escalation)
Parts 1 and 3 will evaluate the ascending dose level of compound 1 in combination with Kang Naifei ni and cetuximab in patients previously treated with BRAF V600E CRC using a rolling six design, irrespective of previous BRAF inhibitor and EGFR inhibitor treatment.
Kang Naifei Ni and 500mg/m in part A1 with 300mg QD 2 The cetuximab combination of Q2W will evaluate compound 1 at multiple QW dose levels of 150 to 250mg (inclusive), such as 150mg QW, 200mg QW and 250mg QW, with 150mg QW as the starting dose.
Kang Naifei Ni and 500mg/m in part A2 with 300mg QD 2 The combination of cetuximab with Q2W will evaluate compound 1 at multiple BID-QW dose levels of 75mg to 125mg (inclusive), such as 75mg BID-QW, 100mg BID-QW and 125mg BID-QW, with 75mg BID-QW as the starting dose.
Kang Naifei Ni and 500mg/m in part A3 with 300mg QD 2 The cetuximab combination of Q2W will evaluate compound 1 at multiple QD dose levels of 20 to 40mg (inclusive), such as 20mg QD, 30mg QD and 40mg QD, with 20mg QD as the starting dose.
After identification of the MID, up to about 15 patients per dose level may be enrolled at MID levels and 1 dose level below the MID to further assess safety, tolerability, and PK/PD. For example, if 200mg QW is part 1 MID, then a 200mg QW group and a 150mg QW group may each recruit a total of about 15 patients (including patients that have been recruited under the rolling six design). The data will be used by the sponsor, negotiating with SRC and researcher to select RD in combination with Kang Naifei ni and cetuximab. The initiator will inform the researcher about the RD choice.
Parts 2 and 4 (dose extension)
In part 2 or part 4 (only one of these parts was selected for expansion), the efficacy of compound 1 plus Kang Naifei ni and cetuximab in patients with BRAF V600E CRC who had not received EC or previous treatment, who had received or had not received previous treatment with BRAF inhibitors and EGFR inhibitors, will be evaluated in multiple steps. Each expanded group in part 2 or part 4 will be subjected to a metaphase analysis to guide the possible early cessation of recruitment in the absence of evidence of activity.
In part 2 or part 4, eligible patients with BRAF V600E CRC will be enrolled in 1 of 2 groups of 2A/4A (patients who have not received prior treatment with BRAF and EGFR inhibitor) or 2B/4B (patients who have received prior treatment with BRAF and EGFR inhibitor).
For either group 2A or 4A, once about 20 evaluable patients enrolled have completed at least 1 post-dose tumor assessment, an interim analysis will be performed in the selected group. If the metaphase analysis of ORR indicates that the antitumor activity is below the threshold for the patient population (e.g., if less than 2 responders are observed in the first 20 patients of the 2A segment), recruitment in the group will cease. Otherwise, up to about 10 additional evaluable patients will be enrolled in the cohort to allow adequate assessment. In addition, up to 70 patients will be enrolled in this cohort for primary efficacy analysis. Safety, PK and PD data will support efficacy analysis and decision to continue recruitment.
For either group 2B or 4B, once about 30 efficacy-evaluable patients of the drug administration have completed at least 1 post-drug tumor assessment, an interim analysis is performed in the selected group. If the metaphase analysis of ORR indicates that the antitumor activity is below the threshold for the patient population (e.g., if no responders are observed in the first 30 evaluable patients in section 2B), recruitment in the population will cease. Otherwise, up to about 70 additional evaluable patients in the cohort will be recruited for primary efficacy assessment. Safety, PK and PD data will support efficacy analysis and decision to continue recruitment.
Dosing regimen
a Compound 1 QW-compound 1 was administered orally once a week;
b compound 1BID-QW-compound 1 was orally administered twice daily during the day of the week;
c compound 1 QD-compound was orally administered once daily;
d EC: 300mg Kang Naifei of the drug is orally taken every day and 500mg/m of the drug is infused intravenously every 2 weeks 2 Cetuximab.
Inclusion criteria:
the age is more than or equal to 18 years old.
Willing and able to give written informed consent.
Based on an analytically validated assay of tumor tissue in a certified test laboratory, there is a histologically or cytologically validated metastatic CRC carrying an applicable mutation (e.g., BRAFm V600E; KRAS or NRAS mutation).
Measurable disease according to the solid tumor response assessment criteria (Response Evaluation Criteria in Solid Tumors, RECIST) v 1.1.
Sufficient bone marrow and organ function.
ECOG performance status with 0 or 1.
Is willing to adhere to all scheme required accesses, evaluations and procedures.
Can swallow oral medicine.
Exclusion criteria:
previous treatments with inhibitors of RAS, MEK or ERK. Other therapies may also be prohibited depending on which treatment group the patient is assigned.
Anticancer treatment of 21 days or less or 4 half-lives (whichever is shorter) before the first dose of drug is studied.
Palliative irradiation for 7 days or less before the first dose of drug was studied.
Symptomatic brain metastasis or leptomeningeal disease.
Gastrointestinal conditions that may affect the absorption of oral medications.
Active infection requiring systemic treatment, or history of HIV, hepatitis b virus or hepatitis c virus infection.
A history of chronic inflammatory bowel disease or Crohn's disease requiring medical intervention (immunomodulating or immunosuppressant drugs or surgery) no more than 12 months prior to the first dose of study drug.
Active, clinically significant interstitial lung disease or pneumonia.
Cardiovascular function is impaired or clinically significant cardiovascular disease.
A history of thromboembolic or cerebrovascular events less than or equal to 6 months prior to the first dose.
Major surgery was anticipated within 28 days of recruitment or during study treatment.
Known intolerance or contraindication to Kang Naifei, cetuximab or palbociclib.
Pregnant or lactating women.
Any evidence of severe or uncontrolled systemic disease or any other significant clinical condition or laboratory discovered evidence that renders the patient unsuitable for participation in the study.
Example 4: 1b/2 phase, open label, multicenter study of Compound 1 in combination with Ornitinib in advanced non-Small cell Lung cancer patients
Detailed description: this is a phase 1b/2, open-label, multicenter clinical study evaluating the combination of compound 1 and octreotide in study participants with advanced NSCLC. This study will be used as a platform study, allowing for evaluation of the safety/tolerability and efficacy of compound 1 in combination with other cancer therapies. In study participants with advanced NSCLC harboring epidermal growth factor receptor sensitizing mutations (EGFRm), the study will initially begin with an ascending dose of compound 1, compound 1 being administered in combination with octenib administered at 80mg once per day (QD) at a dose of 150mg once per week (QW), 200mg QW and 250mg QW, or twice per day once per week (BID-QW), respectively. In study participants with EGFRm NSCLC, dose expansion will be followed and test compound 1 will be administered at the QW Recommended Dose (RD) or BID-QW RD identified from dose escalation in combination with octreotide.
Grouping and intervention
/>
Dosing regimen
/>
Results index
Main result index:
1. dose Limiting Toxicity (DLT). Based on observed adverse events [ time frame: study day 1 to day 29 ].
2. Maximum tolerated dose (MID). Based on observed adverse events [ time frame: study day 1 to day 29 ].
3. Recommended Dose (RD). Based on observed adverse events [ time frame: study day 1 to day 29 ].
4. Adverse events. Incidence and severity of treatment-induced AE and severe AE [ time frame: up to 24 months from the first dose.
Secondary outcome index:
5. plasma concentration (Cmax). Maximum plasma concentrations of compound 1 and other cancer therapies [ time frame: study day 1 to day 29 ].
6. Time to Cmax (Tmax). Time to reach maximum plasma concentrations of compound 1 and other cancer therapies [ time frame: study day 1 to day 29 ].
7. Area under the curve. Area under the plasma concentration-time curve for compound 1 and other cancer therapies [ time range: study day 1 to day 29 ].
8. Half-life period. Half-life of compound 1 and other cancer therapies [ time frame: study day 1 to day 29 ].
9. Objective Response Rate (ORR). Based on the evaluation of radiographic imaging according to RECIST version 1.1 [ time frame: up to 24 months from the first dose.
10. Duration of reaction (DOR). Based on the evaluation of radiographic imaging according to RECIST version 1.1 [ time frame: up to 24 months from the first dose.
Standard: inclusion criteria:
the age is more than or equal to 18 years old.
Willing and able to give written informed consent.
An advanced NSCLC with histological or cytologically confirmed, having EGFR mutations that are sensitive to EGFR inhibitors upon initial diagnosis according to locally approved tags.
Measurable disease according to the solid tumor response assessment criteria (RECIST) v 1.1.
Sufficient bone marrow and organ function.
ECOG performance status with 0 or 1.
Is willing to adhere to all scheme required accesses, evaluations and procedures.
Can be swallowed.
Exclusion criteria:
concurrent with any systemic anti-cancer therapy for NSCLC, including any approved or research agents.
Previous therapies with RAS, RAF, MEK or ERK inhibitors.
Previous Cancer Immunotherapy (CIT) (e.g., immune checkpoint inhibitors) unless a non-CIT containing regimen was followed by CIT prior to study recruitment.
Anticancer treatment was performed within 21 days of recruitment, which may continue during the screening period, except for octreotide.
Palliative radiation treatment was performed within 7 days of recruitment.
A history of unacceptable toxicity to treatment with octenib.
Major surgery was performed within 28 days of enrollment.
Unresolved toxicity from previous systemic therapies was greater than NCI CTCAE.
Grade 1 (except for hair loss) and grade 2 neuropathy at recruitment due to prior chemotherapy.
Any evidence of severe or uncontrolled systemic disease or any other significant clinical condition or laboratory discovered evidence that renders the patient unsuitable for participation in the study.
Cardiovascular function is impaired or clinically significant cardiovascular disease.
Retinal Pigment Epithelial Detachment (RPED), central serous retinopathy, retinal Vein Occlusion (RVO) or historical or current evidence of RPED or RVO-inducing factors.
Pregnant or lactating women.
The contraindication of the use of octreotide in accordance with the local label.
Example 5: in vivo study of Compound 1 alone and Compound 1 in combination with Kang Naifei Nile in the Kang Naifei Nile refractory-BRAF V600E CR0004 PDX model
Vehicle/control preparations were prepared, 100mM acetic acid in deionized water, pH adjusted to 4.8-5.0, and stored at ambient conditions throughout 28 days of administration in mice.
Compound 1 was tested weekly in vehicle of 0.5% CMC and 0.5% tween 80 and stored under ambient conditions. Kang Naifei Ni was prepared weekly in vehicle of 0.5% CMC and 0.5% Tween 80 and stored at 2-8deg.C.
Female Balb/c nude mice were purchased from beijing An Kaiyi boy biotechnology limited (Beijing Anikeeper Biotech co., ltd) (beijing, china). Mice were 9-11 weeks of age at the time of implantation. Mice were housed in an pathogen free (SPF) environment of an animal feeding facility and allowed to adapt to their new environment for at least 3 days before any experiments were initiated according to the IACUC protocol. All procedures related to animal handling, care and treatment in this study were performed according to guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of Crown Bioscience (beijing, china). During the study, care and use of animals was performed according to the guidelines of the laboratory animal care evaluation and approval association (AAALAC). In addition, all parts of this study conducted in Crown Bioscience (beijing, china) followed the study protocol approved by the study owner and applicable Standard Operating Protocol (SOP).
Preparation of PDX
CR0004 PDX model was established at CrownBio for preclinical efficacy studies. The PDX model was derived from 44 year old female CRC patients. The BRAF V600E mutation in this model was confirmed by exome sequencing. The skin of the mice was cleaned on the right abdomen with an appropriate surgical scrub and iodophor. Tumor fragments (2-3 mm diameter) harvested from the PDX model were subcutaneously implanted into the right flank of female BALB/c nude mice using an 18g trocar. When the average tumor size reaches 141mm 3 (in the range of 114-185mm 3 ) At this time, tumor-bearing mice were randomly divided into study groups of 8 mice each.
Treatment of
Treatment was started on the day of randomization. The day of treatment initiation is indicated as day 0 of treatment. Mice were dosed as monotherapy by administration of vehicle control solution, 30 mg/kg/dose of compound 1 of BID p.o. and 90mg/kg of Kang Naifei ni of QD p.o. Another group received 30 mg/kg/dose of BID of Compound 1 in combination with 90mg/kg QD of Kang Naifei Ni. The dosing volume of each compound was 5mL/kg and the BID protocol was 8 hours apart. In the combination group of compound 1 and Kang Naifei ni, kang Naifei ni was administered 1 hour after the first dose of compound 1 BID. The study was terminated on day 28 of treatment as defined in the study protocol.
As shown in fig. 3A, compounds 1 and Kang Naifei ni showed combined in vivo benefits in the Kang Naifei ni refractory BRAF V600E CR0004 PDX model.
Example 6: in vivo study of Compound 1 alone and Compound 1 in combination with Kang Naifei Ni and cetuximab in Kang Naifei Ni+cetuximab refractory BRAF V600E CR0004 PDX model
Vehicle/control preparations were prepared, 100mM acetic acid in deionized water, pH adjusted to 4.8-5.0, and stored at ambient conditions during 28 days of administration in mice.
Test article compound 1 was prepared weekly in a vehicle of 0.5% CMC and 0.5% tween 80 and stored under ambient conditions. Kang Naifei Ni was prepared weekly in vehicle of 0.5% CMC and 0.5% Tween 80 and stored at 2-8deg.C.
Female Balb/c nude mice were purchased from Beijing An Kaiyi Bo Biotechnology Co., ltd (Beijing, china). Mice were 9-11 weeks of age at the time of implantation. Mice were housed in an animal feeding facility in an environment free of Specific Pathogens (SPF) and were acclimatized to the new environment for at least 3 days before any experiments were initiated according to the IACUC protocol. All procedures related to animal handling, care and treatment in this study were performed according to guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of Crown Bioscience (beijing, china). During the study, care and use of animals was performed according to the guidelines of the laboratory animal care evaluation and approval association (AAALAC). In addition, all parts of this study conducted in Crown Bioscience (beijing, china) followed the study protocol approved by the study owner and applicable Standard Operating Protocol (SOP).
Preparation of PDX
CR0004 PDX model was established at CrownBio for preclinical efficacy studies. This PDX model was derived from 44 year old female CRC patients. BRAF in this model was confirmed by exome sequencing V600E Mutation. The skin of the mice was cleaned on the right abdomen with an appropriate surgical scrub and iodophor. Tumor fragments (2-3 mm diameter) harvested from the PDX model were subcutaneously implanted into the right flank of female Balb/c nude mice using an 18g trocar. When the average tumor size reaches 141mm 3 (in the range of 114-185mm 3 ) At this time, tumor-bearing mice were randomly divided into study groups of 8 mice each.
Treatment of
Treatment was started on the day of randomization. The day of treatment initiation is indicated as day 0 of treatment. Mice were dosed as monotherapy by administration of vehicle control solution, 30 mg/kg/dose of compound 1 of BID p.o., kang Naifei ni of QD p.o., 90mg/kg, and 30mg/kg of Q3D i.p. The other three groups received combination treatment, the first group was dosed with 30 mg/kg/dose of compound 1 with 90mg/kg QD Kang Naifei, the second group was dosed with 30 mg/kg/dose of compound 1 with 90mg/kg QD Kang Naifei ni+30 mg/kg Q3D cetuximab, and the third group was dosed with 90mg/kg QD Kang Naifei ni with 30mg/kg Q3D cetuximab. The dosing volume of each compound was 5mL/kg except cetuximab (10 mL/kg), and the BID regimen was 8 hours apart. In the combination group of compound 1 and Kang Naifei ni, kang Naifei ni was administered 1 hour after the first dose of compound 1 BID. In the triple combination group of compound 1 with Kang Naifei and cetuximab, compound 1bid was administered first, kang Naifei ni was administered 1 hour later, and cetuximab was administered 1 hour after Kang Naifei ni. In the Kang Naifei-ni and cetuximab combination group, kang Naifei-ni was administered first and cetuximab was administered 1 hour later. In addition to the conventional food and water supplies, dietGel was added to cages in which at least two mice in the treatment group showed BWL > 10%. According to this practice, mice in the triple combination (i.e., kang Naifei ni of 30mg/kg QD +90mg/kg of compound 1+90mg/kg QD +30mg/kg Q3D cetuximab) were initially supplied with DietGel food on day 21 of treatment and continued for the remaining study period. The study was terminated on day 28 of treatment as defined in the study protocol.
As shown in fig. 3B, the combination of compound 1 with Kang Naifei ni and cetuximab showed in vivo combination benefits in the Kang Naifei ni+cetuximab (EC) refractory BRAF V600E CR0004 PDX model.
Example 7: in vivo study of Compound 1 alone and Compound 1 in combination with Kang Naifei Nile in the Kang Naifei Nile refractory-BRAF V600E CRR1011 PDX model
Vehicle/control preparation of compound 1, 0.5% Methylcellulose (MC) and 0.1% tween 80 in deionized water was prepared and stored at ambient conditions throughout the 23 day administration in mice.
Test article compound 1 was freshly prepared weekly in a vehicle of 0.5% Methylcellulose (MC) and 0.1% tween 80 solution and stored under ambient conditions. The combination agent Kang Naifei Ni was freshly prepared weekly in 0.5% CMC and 0.5% Tween 80 vehicle and stored at 2-8deg.C.
Female Balb/c nude mice were purchased from Beijing Vietnam Lihua laboratory animal technologies Co. Mice were housed in an animal feeding facility in an environment free of Specific Pathogens (SPF) and allowed to adapt to their new environment for at least 3 days before any experiments were initiated. Mice were 6-8 weeks of age at implantation. All procedures related to animal handling, care and treatment in this study were performed according to protocols and guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of genedesign. Animal facilities and procedures operate under the standards of the laboratory animal care and use guidelines (NRC, 2011) and are certified by the laboratory animal care assessment and certification association (AAALAC). Specifically, all parts of the study conducted at genedesign followed the study protocol reviewed and approved by IACUC and the applicable Standard Operating Program (SOP).
Preparation of xenograft models
PDX CRC1011 model was set up in GenenDesign (Shanghai, china) for non-clinical efficacy studies. The PDX model was derived from 63 year old chinese male CRC patient. BRAF in PDX model CRC1011 was confirmed by whole exome sequencing and PCR sequencing V600E Mutation. Tumor fragments harvested from the PDX model were subcutaneously implanted into the right flank of female Balb/c nude mice. Mice were anesthetized with isoflurane and anesthesia was maintained throughout the implantation. The right flank of the mice was sterilized with the appropriate surgical scrub and alcohol and a sterile surgical procedure was used. A small skin incision was made using the sharp end of the trocar and a 1.5cm subcutaneous pocket along the right chest wall was formed by blunt dissection with a 10-12g trocar. Tumor fragments (15-30 mm) 3 ) Placed into a trocar and advanced into a subcutaneous pocket in the right abdomen. The trocar incision is closed with a suture or wound clip and removed one week after closure. When the tumor size reaches 200mm in volume average 3 (in the range of 144-269 mm) 3 ) At this time, tumor-bearing mice were randomly divided into study groups of 8 mice each. Randomization date is expressed as day 0 of treatment.
Treatment of
Treatment was started the following day after randomization. The day of treatment initiation is indicated as day 0 of treatment. Mice were dosed as monotherapy by administration of vehicle control solution, 30 mg/kg/dose of compound 1 of BID p.o. and 90mg/kg of Kang Naifei ni of QD p.o. Another group received 30 mg/kg/dose of BID of Compound 1 in combination with 90mg/kg QD of Kang Naifei Ni. The dosing volumes of compound 1 and Kang Naifei Ni were 5mL/kg with 8 hours intervals for the BID protocol. In the combination group of compounds 1 and Kang Naifei, kang Naifei of the second was administered first for 1 hour The first dose of compound 1BID was then administered. In addition to the conventional food and water supply, at least two mice therein showed>Dietgel was added to 10% BWL cages. According to this practice, mice in the 30 mg/kg/dose of compound 1 monotherapy group of BID and the 30 mg/kg/dose of compound 1 in combination with Kang Naifei of QD 90mg/kg of BID were supplied with DietGel food, starting on days 12, 12 and 9 of treatment, respectively, and continued for the remaining study period. Due to rapid tumor growth in the vehicle group, the study terminated on day 23 of treatment, earlier than the original termination day (day 28) as defined in the study protocol. On day 23 of treatment, two of the eight tumors in the vehicle control group exceeded the tumor volume threshold (2,000 mm 3 )。
As shown in fig. 4A, compounds 1 and Kang Naifei ni showed combined in vivo benefits in the Kang Naifei ni refractory BRAF V600E CRC1011 PDX model.
Example 8: in vivo study of Compound 1 alone and Compound 1 in combination with Kang Naifei Ni and cetuximab in Kang Naifei Ni+cetuximab refractory BRAF V600E CRC1011 PDX model
Test article compound 1 was manufactured by WuXi STA in Shanghai, china. The combination agent Kang Naifei is available from MedChemexpress. Cetuximab was purchased from Merck KGaA (lot G002 VX) in china.
Vehicle/control preparation of compound 1, 0.5% Methylcellulose (MC) and 0.1% tween 80 in deionized water was prepared and stored at ambient conditions throughout the 23 day administration in mice.
Test article compound 1 was freshly prepared weekly in a vehicle of 0.5% Methylcellulose (MC) and 0.1% tween 80 solution and stored under ambient conditions. The combination agent Kang Naifei Ni was freshly prepared weekly in 0.5% CMC and 0.5% Tween 80 vehicle and stored at 2-8deg.C. Prior to each administration, cetuximab (5 mg/mL) was diluted to 3mg/mL with saline.
Female Balb/c nude mice were purchased from Beijing Vietnam Lihua laboratory animal technologies Co. Mice were housed in an animal feeding facility in an environment free of Specific Pathogens (SPF) and allowed to adapt to their new environment for at least 3 days before any experiments were initiated. Mice were 6-8 weeks of age at implantation. All procedures related to animal handling, care and treatment in this study were performed according to protocols and guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of genedesign. Animal facilities and procedures operate under the standards of the laboratory animal care and use guidelines (NRC, 2011) and are certified by the laboratory animal care assessment and certification association (AAALAC). Specifically, all parts of the study conducted at genedesign followed the study protocol reviewed and approved by IACUC and the applicable Standard Operating Program (SOP).
Preparation of PDX
PDX CRC1011 model was set up in GenenDesign (Shanghai, china) for non-clinical efficacy studies. The PDX model was derived from 63 year old chinese male CRC patient. BRAF in PDX model CRC1011 was confirmed by whole exome sequencing and PCR sequencing V600E Mutation. Tumor fragments harvested from the PDX model were subcutaneously implanted into the right flank of female Balb/c nude mice. Mice were anesthetized with isoflurane and anesthesia was maintained throughout the implantation. The right flank of the mice was sterilized with the appropriate surgical scrub and alcohol and a sterile surgical procedure was used. A small skin incision was made using the sharp end of the trocar and a 1.5cm subcutaneous pocket along the right chest wall was formed by blunt dissection with a 10-12g trocar. Tumor fragments (15-30 mm) 3 ) Placed into a trocar and advanced into a subcutaneous pocket in the right abdomen. The trocar incision is closed with a suture or wound clip and removed one week after closure. When the tumor size reaches 200mm in volume average 3 (in the range of 144-269 mm) 3 ) At this time, tumor-bearing mice were randomly divided into study groups of 8 mice each. Randomization date is expressed as day 0 of treatment.
Treatment of
Treatment was started the following day after randomization. The day of treatment initiation is indicated as day 0 of treatment. Mice were dosed as monotherapy by administration of vehicle control solution, 30 mg/kg/dose of compound 1 of BID p.o., kang Naifei ni of QD p.o., 90mg/kg, and 30mg/kg of Q3D i.p. Three additional groups were treated with the combination, the first group was dosed with 30 mg/kg/dose of Compound 1 of BID and Kang Naifei Ni of 90mg/kg QD, the second group was dosed with 30mg Compound 1 per kg/dose BID was administered with Kang Naifei Ni of 90mg/kg QD+30 mg/kg Q3D cetuximab, and the third group was administered with Kang Naifei Ni of 90mg/kg QD and 30mg/kg Q3D cetuximab. The dosing volumes of compound 1, kang Naifei and cetuximab were 5mL/kg with 8 hour intervals of the BID regimen. In the combination group of compound 1 and Kang Naifei ni, kang Naifei ni was administered first and 1 hour later the first dose of compound 1BID was administered. In the combination group of compound 1 with Kang Naifei ni and cetuximab, kang Naifei ni was administered first, the first dose of compound 1BID was administered after 1 hour, and cetuximab was administered 1 hour after the first dose of compound 1 BID. In the Kang Naifei-ni and cetuximab combination group, kang Naifei-ni was administered first and cetuximab was administered 1 hour later. In addition to the conventional food and water supply, at least two mice therein showed>Dietgel was added to 10% BWL cages. According to this practice, mice in the 30 mg/kg/dose of compound 1 monotherapy group of BID, the 30 mg/kg/dose of compound 1 in combination with Kang Naifei of QD and the 30 mg/kg/dose of compound 1 in combination with Kang Naifei of qd+30 mg/kg of Q3D cetuximab of QD were supplied with DietGel food starting on days 12, 12 and 9 of treatment, respectively, and continued for the remaining study period. Because of the rapid growth of tumors in the vehicle group, the study terminated on day 23 of treatment, earlier than the original termination day defined in the study protocol (day 28). On day 23 of treatment, two of the eight tumors in the vehicle control group exceeded the tumor volume threshold (2,000 mm 3 )。
As shown in fig. 4B, the combination of compound 1 with Kang Naifei ni and cetuximab showed in vivo combined benefits in the Kang Naifei ni+cetuximab (EC) refractory BRAF V600E CRC1011 PDX model.

Claims (135)

1. A method of treating cancer in a subject in need thereof, the method comprising: administering to said subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof, and
(ii) EGFR inhibitors.
2. The method of claim 1, wherein the EGFR inhibitor is a small molecule EGFR inhibitor.
3. The method of claim 1, wherein the EGFR inhibitor is afatinib, angstrom Mo Tuoshan antibody, canetinib, cetuximab, dactyltinib, daphnetin, erlotinib, gefitinib, icotinib, lapatinib, lazetidine, liffenib, crizotuzumab, mo Bo tinib, natatinib, cetuximab, lenatinib, octyitinib, panitumumab, pelitinib, wave Ji Tini, tivozanib, rocitinib, sapitinib, vandetanib, or valitinib.
4. The method of any one of claims 1 to 3, wherein the EGFR inhibitor is octenib.
5. The method of claim 4, wherein the octreotide is administered in an amount of about 80 mg/day.
6. The method of claim 1, wherein the EGFR inhibitor is cetuximab.
7. The method of claim 6, wherein cetuximab is initially at 400mg/m once a week 2 Applied over 120 minutes, then at 250mg/m 2 Applied over a period of 60 minutes.
8. The method of claim 6, wherein cetuximab is administered at 500mg/m once every two weeks 2 And (3) application.
9. The method of claim 6, wherein cetuximab is administered at 400mg/m once every two weeks 2 Administered or once every two weeks at 300mg/m 2 And (3) application.
10. The method of any one of claims 1-9, wherein the method further comprises administering a BRAF inhibitor.
11. The method of claim 10, wherein the BRAF inhibitor is dabrafenib, kang Naifei ni, regorafenib, sorafenib, or vemurafenib.
12. The method of claim 10, wherein the BRAF inhibitor is Kang Naifei ni.
13. The method of claim 12, wherein Kang Naifei ni is administered in an amount of about 100 mg/day to about 500 mg/day.
14. The method of claim 12, wherein Kang Naifei ni is administered in an amount of about 450 mg/day.
15. The method of claim 12, wherein Kang Naifei ni is administered in an amount of about 300 mg/day.
16. The method of claim 12, wherein Kang Naifei ni is administered in an amount of about 225 mg/day.
17. The method of claim 12, wherein Kang Naifei ni is administered in an amount of about 150 mg/day.
18. The method of claim 10, wherein the BRAF inhibitor is dabrafenib.
19. The method of claim 18, wherein dabrafenib is administered in an amount of about 150 mg.
20. A method of treating cancer in a subject in need thereof, the method comprising: administering to said subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) An EGFR inhibitor; and
(iii) BRAF inhibitors.
21. The method of claim 20, wherein the EGFR inhibitor is a small molecule inhibitor.
22. The method of claim 20, wherein the EGFR inhibitor is afatinib, an angstrom Mo Tuoshan antibody, cetuximab, dactyltinib, erlotinib, gefitinib, lapatinib, lasfitinib, lifenib, crizotinib, mo Bo tinib, natatinib, cetuximab, lenatinib, octreotide, or vandetanib.
23. The method of any one of claims 20-22, wherein the EGFR inhibitor is octenib.
24. The method of claim 23, wherein the octreotide is administered in an amount of about 80 mg/day.
25. The method of claim 22, wherein the EGFR inhibitor is cetuximab.
26. The method of claim 25, wherein cetuximab is initially at 400mg/m once a week 2 Applied over 120 minutes, then at 250mg/m 2 Applied over a period of 60 minutes.
27. The method of claim 25, wherein cetuximab is administered at 500mg/m once every two weeks 2 And (3) application.
28. The method according to claim 25, which comprisesThe content of the cetuximab in the composition is 400mg/m once every two weeks 2 Administered or once every two weeks at 300mg/m 2 And (3) application.
29. The method of any one of claims 20-28, wherein the BRAF inhibitor is dabrafenib, kang Naifei ni, regorafenib, sorafenib, or vemurafenib.
30. The method of any one of claims 20-29, wherein the BRAF inhibitor is Kang Naifei ni.
31. The method of claim 30, wherein Kang Naifei ni is administered in an amount of about 100 mg/day to about 500 mg/day.
32. The method of claim 30, wherein Kang Naifei ni is administered in an amount of about 450 mg/day.
33. The method of claim 30, wherein Kang Naifei ni is administered in an amount of about 300 mg/day.
34. The method of claim 30, wherein Kang Naifei ni is administered in an amount of about 225 mg/day.
35. The method of claim 30, wherein Kang Naifei ni is administered in an amount of about 150 mg/day.
36. The method of any one of claims 20-29, wherein the BRAF inhibitor is dabrafenib.
37. The method of claim 36, wherein dabrafenib is administered in an amount of about 150 mg/day.
38. A method of treating cancer in a subject in need thereof, the method comprising: administering to said subject in need thereof a therapeutically effective amount of
(i) Chemical combinationObject 1:or a pharmaceutically acceptable salt thereof,
(ii) Cetuximab; and
(iii) Kang Naifei Ni.
39. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Cetuximab; and
(iii) Darafenib.
40. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Ornitinib; and
(iii) Kang Naifei Ni.
41. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject in need thereof a therapeutically effective amount of
(i) Compound 1:or a pharmaceutically acceptable salt thereof,
(ii) Ornitinib; and
(iii) Darafenib.
42. The method according to any one of claims 1-41, wherein the pharmaceutically acceptable salt of compound 1 is mandelate.
43. The method of any one of claims 1-42, wherein the cancer is a mitogen-activated protein kinase (MAPK) pathway-driven cancer.
44. The method according to any one of claims 1-42, wherein the cancer is BRAF-driven cancer, HRAS-driven cancer, or NRAS-driven cancer.
45. The method of any one of claims 1-42, wherein the cancer comprises at least one cancer cell driven by deregulated ERK.
46. The method of any one of claims 1-42, wherein the cancer has at least one mutation in the RAS.
47. The method of any one of claims 1-42, wherein the cancer has at least one mutation in RAF.
48. The method of any one of claims 1-42, wherein the cancer has at least one mutation in MEK.
49. The method of any one of claims 1-42, wherein the cancer has a G12C KRAS mutation.
50. The method of any one of claims 1-42, wherein the cancer has a G12D KRAS mutation.
51. The method of any one of claims 1-42, wherein the cancer has a G12S KRAS mutation.
52. The method of any one of claims 1-42, wherein the cancer has a G12V KRAS mutation.
53. The method of any one of claims 1-42, wherein the cancer has a G13D KRAS mutation.
54. The method of any one of claims 1-42, wherein the cancer has a Q16H KRAS mutation.
55. The method of any one of claims 1-42, wherein the cancer has a Q16K KRAS mutation.
56. The method of any one of claims 1-42, wherein the cancer has a Q61R NRAS mutation.
57. The method according to any one of claims 1-42, wherein the cancer is a BRAF V600E or V600K mutant tumor.
58. The method of any one of claims 1-42, wherein the cancer is a pan-carcinoma that does not receive MAPKm/MAPKi.
59. The method of any one of claims 1-42, wherein the cancer comprises one or more EGFR mutations selected from the group consisting of: increased EGFR gene copy, EGFR gene amplification, chromosome 7 polyhedra, L858R, exon 19 deletion/insertion, L861Q, G719C, G719S, G719A, V765A, T783A, exon 20 insertion, EGFR splice variants (Viii, vvi and Vii), A289D, A289T, A289V, G598A, G598V, T790M and C797S.
60. The method of any one of claims 1-42, wherein the cancer comprises one or more EGFR mutations selected from the group consisting of L858R, exon 19 deletion, and T790M.
61. The method of any one of claims 1-60, wherein the cancer is a solid tumor.
62. The method of any one of claims 1-61, wherein the cancer is non-small cell lung cancer (NSCLC), melanoma, pancreatic cancer, salivary gland tumor, thyroid cancer, colorectal cancer (CRC), or esophageal cancer.
63. The method of any one of claims 1-62, wherein the cancer is non-small cell lung cancer (NSCLC).
64. The method of claim 63, wherein the NSCLC is an EGFR mutant NSCLC.
65. The method of claim 63, wherein the NSCLC is a KRAS G12C mutant NSCLC.
66. The method of claim 63, wherein the NSCLC is a KRAS G12D mutant NSCLC.
67. The method of claim 63, wherein the NSCLC is KRAS G12S mutant NSCLC.
68. The method of claim 63, wherein the NSCLC is KRAS G12V mutant NSCLC.
69. The method of claim 63, wherein the NSCLC is KRAS G13D mutant NSCLC.
70. The method of claim 63, wherein the NSCLC is a KRAS Q61H mutant NSCLC.
71. The method of claim 63, wherein the NSCLC is KRAS Q61K mutant NSCLC.
72. The method of claim 63, wherein the NSCLC is NRAS Q61R mutant NSCLC.
73. The method of claim 63, wherein the cancer is NSCLC that does not receive MAPKm/MAPKi.
74. The method of claim 63, wherein the cancer is BRAFi treated V600 NSCLC.
75. The method of claim 63, wherein the cancer is KRAS-treated G12C NSCLC.
76. The method of claim 63, wherein the cancer is KRAS-treated G12D NSCLC.
77. The method of claim 63, wherein the cancer is KRAS-treated G12S NSCLC.
78. The method of claim 63, wherein the cancer is KRAS-treated G12V NSCLC.
79. The method of claim 63, wherein the cancer is KRAS-treated G13D NSCLC.
80. The method of claim 63, wherein the cancer is KRAS treated Q61H NSCLC.
81. The method of claim 63, wherein the cancer is KRAS treated Q61K NSCLC.
82. The method of claim 63, wherein the cancer is NRAS treated Q61R NSCLC.
83. The method of any one of claims 1-82, wherein the cancer is pancreatic cancer.
84. The method of claim 83, wherein the cancer is pancreatic cancer that does not receive MAPKm/MAPKi.
85. The method of any one of claims 1-62, wherein the cancer is melanoma.
86. The method of claim 85, wherein the melanoma is BRAF V600E or V600K mutant tumor.
87. The method of claim 85, wherein the cancer is BRAFi treated V600 melanoma.
88. The method of any one of claims 1-62, wherein the cancer is a salivary gland tumor.
89. The method of any one of claims 1-62, wherein the cancer is thyroid cancer.
90. The method of any one of claims 1-62, wherein the cancer is colorectal cancer (CRC).
91. The method of claim 90, wherein the CRC is BRAF V600ECRC.
92. The method of claim 90, wherein the CRC is a KRAS mutant CRC.
93. The method of claim 90, wherein the CRC is a KRAS G12C mutant CRC.
94. The method of claim 90, wherein the CRC is a KRAS G12D mutant CRC.
95. The method of claim 90, wherein the CRC is a KRAS G12S mutant CRC.
96. The method of claim 90, wherein the CRC is a KRAS G12V mutant CRC.
97. The method of claim 90, wherein the CRC is a KRAS G13D mutant CRC.
98. The method of claim 90, wherein the CRC is a KRAS Q61H mutant CRC.
99. The method of claim 90, wherein the CRC is a KRAS Q61K mutant CRC.
100. The method of claim 90, wherein the CRC is an NRAS mutant CRC.
101. The method of claim 100, wherein the CRC is an NRAS Q61R mutant CRC.
102. The method of any one of claims 1-62, wherein the cancer is esophageal cancer.
103. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25 mg/day to about 300 mg/day.
104. The method of any one of claims 1-103, wherein compound 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 25 mg/day to 150 mg/day.
105. The method of any one of claims 1-104, wherein compound 1 or a pharmaceutically acceptable salt thereof is administered in an amount of about 25 mg/day, about 50 mg/day, about 75 mg/day, about 100 mg/day, about 125 mg/day, about 150 mg/day, about 175 mg/day, about 200 mg/day, about 225 mg/day, or about 250 mg/day.
106. The method of any one of claims 1-105, wherein compound 1 or a pharmaceutically acceptable salt thereof is administered in an amount of about 25 mg/day, about 50 mg/day, about 100 mg/day, or about 150 mg/day.
107. The method of any one of claims 1-105, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 250 mg/day.
108. The method of any one of claims 1-107, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered once daily (QD).
109. The method of any one of claims 1-107, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered twice daily (BID).
110. The method of any one of claims 1-107, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered three times per day (TID).
111. The method of any one of claims 1-110, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered weekly.
112. The method of any one of claims 1-110, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered twice weekly.
113. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 300mg twice daily, one day per week (BID-QW).
114. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 250mg twice daily, one day per week (BID-QW).
115. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg to about 150mg twice daily, one day per week (BID-QW).
116. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 50mg, about 75mg, about 100mg, about 125mg, about 150mg, about 175mg, about 200mg, about 225mg, or about 250mg twice daily, one day per week (BID-QW).
117. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 25mg, 50mg, about 100mg, about 125mg, or about 150mg twice daily, one day per week (BID-QW).
118. The method of any one of claims 1-102, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered in an amount of about 125mg twice daily, one day per week (BID-QW).
119. The method of any one of claims 1-118, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered for at least one 28-day period.
120. The method of any one of claims 1-119, wherein compound 1 or a pharmaceutically acceptable salt thereof is administered on days 1, 8, 15, and 22 of a 28 day cycle.
121. The method of any one of claims 1-119, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered on day 1, day 8, day 15 of a 28 day cycle.
122. The method of any one of claims 1-120, wherein compound 1, or a pharmaceutically acceptable salt thereof, is administered orally.
123. The method of any one of claims 1-121, wherein the method further comprises administering an additional MAPK pathway inhibitor.
124. The method of claim 123, wherein the additional MAPK pathway inhibitor is a KRAS inhibitor, NRAS inhibitor, HRAS inhibitor, PDGFRA inhibitor, PDGFRB inhibitor, MET inhibitor, FGFR inhibitor, ALK inhibitor, ROS1 inhibitor, TRKA inhibitor, TRKB inhibitor, TRKC inhibitor, EGFR inhibitor, IGFR1R inhibitor, GRB2 inhibitor, SOS inhibitor, ARAF inhibitor, BRAF inhibitor, RAF1 inhibitor, MEK2 inhibitor, c-Mycv, CDK4/6, inhibitor CDK2 inhibitor, FLT3 inhibitor, or ERK1/2 inhibitor.
125. The method of claim 123, wherein the additional MAPK pathway inhibitor is a KRAS inhibitor.
126. The method of claim 123, wherein the additional MAPK pathway inhibitor is a BRAF inhibitor.
127. The method of claim 123, wherein the additional MAPK pathway inhibitor is an EGFR inhibitor.
128. The method of claim 123, wherein the additional MAPK pathway inhibitor is CDK4/6.
129. The method of claim 123, wherein the additional MAPK pathway inhibitor is a FLT3 inhibitor.
130. The method of claim 123, wherein the additional MAPK pathway inhibitor is adaglabra, afatinib, ASTX029, bimatinib, cobicitinib, dacatinib, erlotinib, gefitinib, ji Ruiti, lapatinib, LTT462, LY3214996, rituximab, lenatinib, nimotuzumab, pamazenib, sematinib, sotolsibirib, trimatinib, ulitinib, vandetanib, or valrfenib.
131. The method of claim 123, wherein the additional MAPK pathway inhibitor is adaglazeb.
132. The method of claim 123, wherein the additional MAPK pathway inhibitor is gefitinib.
133. The method of claim 123, wherein the additional MAPK pathway inhibitor is palbociclib.
134. The method of claim 123, wherein the additional MAPK pathway inhibitor is panitumumab.
135. The method of claim 123, wherein the additional MAPK pathway inhibitor is sotoraciclovir.
CN202280057485.0A 2021-06-24 2022-06-23 ERK1/2 and EGFR inhibitor combination therapies Pending CN117858725A (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US63/214,765 2021-06-24
US63/236,635 2021-08-24
US63/277,547 2021-11-09
US63/279,877 2021-11-16
US202263321605P 2022-03-18 2022-03-18
US63/321,605 2022-03-18
PCT/US2022/034660 WO2022271907A1 (en) 2021-06-24 2022-06-23 Erk1/2 and egfr inhibitors combination therapy

Publications (1)

Publication Number Publication Date
CN117858725A true CN117858725A (en) 2024-04-09

Family

ID=90529096

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280057485.0A Pending CN117858725A (en) 2021-06-24 2022-06-23 ERK1/2 and EGFR inhibitor combination therapies

Country Status (1)

Country Link
CN (1) CN117858725A (en)

Similar Documents

Publication Publication Date Title
US12011449B2 (en) Therapeutic combinations comprising a c-RAF inhibitor
US9486445B2 (en) Combination therapy for proliferative disorders
US8709419B2 (en) Combination therapy
CN110494166A (en) Combination treatment
US20240299388A1 (en) Erk1/2 and shp2 inhibitors combination therapy
US20240285625A1 (en) Erk1/2 and kras g12c inhibitors combination therapy
JP2024514202A (en) Use of heterocyclic inhibitors of ERK1/2
CN117858725A (en) ERK1/2 and EGFR inhibitor combination therapies
US20240307392A1 (en) Erk1/2 and egfr inhibitors combination therapy
CN117897158A (en) ERK1/2 and SHP2 inhibitor combination therapies
WO2022271939A1 (en) Erk1/2 and cdk4/6 inhibitors combination therapy
US20240316046A1 (en) Erk1/2 inhibitor combination therapy
CA3223692A1 (en) Erk1/2 inhibitor combination therapy
CN117940132A (en) ERK1/2 and KRAS G12C inhibitor combination therapies
TW202416986A (en) Methods of treating disorders with ulk inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination