CN117396233A - Combination of MCL-1 inhibitors with antibody drug conjugates - Google Patents

Combination of MCL-1 inhibitors with antibody drug conjugates Download PDF

Info

Publication number
CN117396233A
CN117396233A CN202280038962.9A CN202280038962A CN117396233A CN 117396233 A CN117396233 A CN 117396233A CN 202280038962 A CN202280038962 A CN 202280038962A CN 117396233 A CN117396233 A CN 117396233A
Authority
CN
China
Prior art keywords
cancer
antibody
compound
administered
inhibitor
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
CN202280038962.9A
Other languages
Chinese (zh)
Inventor
T·F·肯尼
C·K·马特森
C·文卡塔拉马尼
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.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Priority claimed from PCT/US2022/032816 external-priority patent/WO2022261310A1/en
Publication of CN117396233A publication Critical patent/CN117396233A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure relates generally to methods of treating cancer by administering an MCL-1inhibitor and an antibody-drug conjugate.

Description

Combination of MCL-1 inhibitors with antibody drug conjugates
Cross Reference to Related Applications
The present application claims the benefit of U.S. provisional application Ser. No. 63/209,667 filed on day 11, 6, 2021 and U.S. provisional application Ser. No. 63/322,509 filed on day 22, 3, 2022. The entire contents of these applications are incorporated herein by reference in their entirety.
Technical Field
The present application relates generally to combination therapies of MCL-1 inhibitors and antibody-drug conjugates (ADCs) for the treatment of cancer. In particular, the antibody is an anti-Trop-2 antibody and the drug is an anticancer agent.
Background
Apoptosis (programmed cell death) is the process of eliminating unwanted or potentially dangerous cells from an organism. Avoiding apoptosis is critical for the development and continued growth of tumors. Myeloid leukemia 1 protein (MCL-1, also abbreviated as MCL-1 or MCL 1) is an anti-apoptotic member of the Bcl-2 protein family. MCL-1 is overexpressed in many cancers. Overexpression of MCL-1 prevents cancer cells from undergoing apoptosis.
Studies have shown that MCL-1 inhibitors are useful in the treatment of a variety of cancers. See, e.g., the MCL1inhibitor S63845 is tolerable and effective in diverse cancer models A.Kotschy et al, nature,2016 (538): 477-482; "Structure Based Design of Non-Natural Peptidic Macrocyclic Mcl-1 Inhibitors"J.Johannes et al, ACS Med. Chem. Lett.,2017,8 (2): 239-244&ACS Med.Chem.Lett, 2017,8 (11): 1204; "Synergistic action of the MCL-1inhibitor S63845 with current therapies in preclinical models of triple-negative and HER2-amplified breast cancer" D.Merno et al, sci.Transl.Med., month 8, day 2, 2017, 9 (401): eaam7049; "Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia" A.Tron et al, nature Comm.2018 (9): article No.5341; "AMG 176,a Selective MCL1Inhibitor,Is Effective in Hematologic Cancer Models Alone and in Combination with Established Therapies"S.Caenepeel et al, cancer discover., 2018, month 12 (12): 1582-1597; "Discovery of S64315, a Potent and Selective Mcl-1inhibitor" Z.Szlavik et al, J.Med.chem.,2020,63 (22): 13762-13795.
In recent years, ADCs comprising tumor-associated monoclonal antibodies (mabs) and anti-cancer agents have been developed in cancer therapies. For example, the salcetuximab goverikang (sacituzumab govitecan) disclosed in U.S. patent No. 7,999,083 comprises Trop-2 antibody and SN-38.
There remains a need to provide more effective methods for treating cancer.
Disclosure of Invention
In some embodiments, provided herein is a method of treating cancer, the method comprising: administering to a human patient in need thereof a therapeutically effective amount of an antibody-drug conjugate and a therapeutically effective amount of an MCL-1 inhibitor;
wherein the antibody-drug conjugate comprises an anti-Trop-2 antibody and an anti-cancer agent; and wherein the MCL-1 inhibitor has formula (I) or a pharmaceutically acceptable salt thereof:
wherein R is 1 Is a 5-10 membered heteroaryl group containing 1-2 heteroatoms; wherein each heteroatom is independently selected from nitrogen, sulfur, and oxygen;
R 1 optionally independently selected from halogen, hydroxy, -CN, C 1-6 Alkyl, C 1-6 Haloalkyl, -OR a And C 3-6 1-3 substituents of cycloalkyl; and is also provided with
Each R 2 、R 3 、R 4 And R is 5 Independently hydrogen or C 1-6 An alkyl group;
R 6 hydrogen or halogen; and is also provided with
R a Independently hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl and C 3-10 Cycloalkyl groups.
Drawings
FIG. 1 paclitaxel treatment increased FBXW7 protein and decreased MCL1 protein and MCL1-BAK and MCL1-BIM protein dimers in TNBC cells.
Figure 2HCC70 inhibition and 95% CI synergistic response surface.
FIG. 3MDA-MB-468 inhibition and 95% CI synergistic response surface.
Figure 4HCC1806 inhibition and 95% CI synergistic response surface.
FIG. 5TNBC PDX model CTG-1909 tumor growth.
FIG. 6TNBC PDX model CTG-2010 tumor growth.
FIG. 7MDA-MB-468 tumor growth.
Detailed Description
Definition of the definition
Throughout the specification and claims, unless the context requires otherwise, the word "comprise" and variations such as "comprises" and "comprising" will be interpreted in an open, inclusive sense, i.e. as "including but not limited to".
Prefix such as "C u-v Or (C) u -C v ) Indicating that the following groups have u to v carbon atoms, where u and v are integers. For example, "C 1-6 Alkyl "indicates that the alkyl group has 1 to 6 carbon atoms.
A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -C (O) NH 2 Through a carbon atom. Dashes at the front or end of the chemical group are for convenience; chemical groups can be depicted without one or more dashes without losing their ordinary meaning. No directionality is indicated or implied by the order in which chemical groups are written or named unless chemical or structural requirements.
The term "substituted" means that one or more hydrogen atoms on the hydrocarbon is replaced with one or more atoms or groups other than hydrogen, provided that the normal valency of the designated carbon atom(s) is not exceeded. "substituents" are atoms or groups that replace a hydrogen atom when a hydrocarbon is "substituted". Unless otherwise indicated, where groups are described as optionally substituted, any substituents of these groups are themselves unsubstituted.
Reference herein to "about" a value or parameter includes (and describes) embodiments that relate to the value or parameter itself. In certain embodiments, the term "about" includes the indicated amount ± 10%. In other embodiments, the term "about" includes the indicated amount ± 5%. In certain other embodiments, the term "about" includes the indicated amount ± 1%. Moreover, the term "about X" includes descriptions of "X". Moreover, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a compound" includes a plurality of such compounds, and reference to "an assay" includes reference to one or more assays known to those skilled in the art and equivalents thereof.
"alkyl" refers to an unbranched or branched saturated hydrocarbon chain. As used herein, alkyl groups have 1 to 20 carbon atoms (i.e., C 1-20 Alkyl), 1 to 12 carbon atoms (i.e., C 1-12 Alkyl), 1 to 8 carbon atoms (i.e., C 1-8 Alkyl), 1 to 6 carbon atoms (i.e., C 1-6 Alkyl), 1 to 4 carbon atoms (i.e., C 1-4 Alkyl), 1 to 3 carbon atoms (i.e., C 1-3 Alkyl) or 1 to 2 carbon atoms (i.e., C 1-2 Alkyl). Examples of alkyl groups include, but are not limited to: methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl and 3-methylpentyl. When an alkyl residue having a particular carbon number is named by chemical name or identified by molecular formula, all positional isomers having that carbon number are contemplated; thus, for example, a "butyl" includes n-butyl (i.e., - (CH) 2 ) 3 CH 3 ) Sec-butyl (i.e. -CH (CH) 3 )CH 2 CH 3 ) Isobutyl (i.e. -CH) 2 CH(CH 3 ) 2 ) And tert-butyl (i.e. -C (CH) 3 ) 3 ) The method comprises the steps of carrying out a first treatment on the surface of the And "propyl" includes n-propyl (i.e. (CH) 2 ) 2 CH 3 ) And isopropyl (i.e. -CH (CH) 3 ) 2 )。
"aryl" refers to an aromatic carbocyclic group having a single ring (e.g., monocyclic) or multiple rings (e.g., bicyclic or tricyclic) containing fused systems. As used herein, aryl has 6 to 20 ring carbon atoms (i.e., C6-20 aryl), 6 to 12 carbon ring atoms (i.e., C6-12 aryl), or 6 to 10 carbon ring atoms (i.e., C6-10 aryl). Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, and anthracyl. However, aryl does not encompass or overlap in any way with heteroaryl as defined below. If one or more aryl groups are fused to a heteroaryl ring, the resulting ring system is heteroaryl.
"cycloalkyl" refers to a saturated or partially saturated cyclic alkyl group having a single ring or multiple rings including fused, bridged, and spiro ring systems. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
"halo" and "halogen" are used herein to refer to fluorine (-F), chlorine (-Cl), bromine (-Br) and iodine (-I).
As used herein, the term "haloalkyl" refers to an alkyl group as defined herein wherein one or more hydrogen atoms of the alkyl group are independently substituted with the same or different halogen substituents. For example, C 1-6 Haloalkyl is C 1-6 Alkyl, wherein C 1-6 One or more of the hydrogen atoms of the alkyl group have been substituted with a halo substituent. Examples of haloalkyl groups include, but are not limited to, fluoromethyl, fluorochloromethyl, difluoromethyl, difluorochloromethyl, trifluoromethyl, 1-trifluoroethyl and pentafluoroethyl.
"heteroaryl" refers to an aromatic group, including groups having an aromatic tautomer or resonance structure, having a single, multiple, or multiple fused rings, which have at least one heteroatom in the ring, i.e., one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein either nitrogen or sulfur may be oxidized. Thus, the term includes a ring having one or more of O, N, S, S (O), S (O) 2 And a ring of N-oxide groups. The term includes rings having one or more cyclic C (O) groups. As used herein, heteroaryl includes 5 to 20 ring atoms (i.e., 5 to 20 membered heteroaryl), 5 to 12 ring atoms (i.e., 5 to 12 membered heteroaryl), or 5 to 10 ring atoms (i.e., 5 to 10 membered heteroaryl) and 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur, as well as oxidized forms of the heteroatoms. Examples of heteroaryl groups include pyridin-2 (1H) -one, pyridazin-3 (2H) -one, pyrimidin-4 (3H) -one, quinolin-2 (1H) -one, pyrimidinyl, purinyl, pyridinyl, pyridazinyl, benzothiazolyl and pyrazolyl. Heteroaryl does not encompass or overlap with aryl as defined above.
The term "heterocyclyl", "heterocycle" means having a single ring or multiple ringsA mono-or di-radical saturated or unsaturated group of fused rings having one or more heteroatoms selected from nitrogen, sulfur, phosphorus and/or oxygen within the ring. The heteroatoms within the "heterocyclyl" may be oxidized, for example, -N (O) -, -S (O) - 2 -. The heterocyclyl may be a single ring or multiple rings, wherein the multiple rings may be fused, bridged or spiro.
"isomers" are different compounds having the same molecular formula. Isomers include stereoisomers, enantiomers and diastereomers.
"stereoisomers" refers to compounds that consist of the same atoms bonded by the same bonds but have different three-dimensional structures that are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof, and includes "enantiomers," which refers to two stereoisomers whose molecules are non-superimposable mirror images of each other.
"tautomer" refers to proton transfer from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any of the compounds.
"solvates" are formed by the interaction of a solvent and a compound. Solvates of salts of the compounds described herein are also provided. Hydrates of the compounds described herein are also provided.
As used herein, the term "prodrug" is a biologically inactive derivative of a drug that is converted to the biologically active parent drug following administration to the human body according to some chemical or enzymatic route.
"enantiomers" are a pair of stereoisomers that are non-superimposable mirror images of each other. The 1:1 mixture of a pair of enantiomers is a "racemic" mixture. The symbol "(±)" is used to denote a racemic mixture where appropriate.
"diastereomers" are stereoisomers that have at least two asymmetric atoms but which are not mirror images of each other.
As used herein, "treatment" or "treatment" is a method for achieving a beneficial or desired result. For purposes of this disclosure, beneficial or desired results include, but are not limited to, alleviation of symptoms and/or diminishment of extent of symptoms associated with a disease or condition. In one embodiment, "treatment" or "treatment" includes one or more of the following: a) Inhibiting the disease or disorder (e.g., reducing one or more symptoms caused by the disease or disorder, and/or reducing the extent of the disease or disorder); b) Slowing or arresting the development of one or more symptoms associated with the disease or disorder (e.g., stabilizing the disease or disorder, delaying the progression or worsening of the disease or disorder); and c) alleviating the disease or condition, e.g., causing regression of clinical symptoms, ameliorating the disease state, delaying the progression of the disease, improving quality of life, and/or prolonging survival.
As used herein, "prevention" or "prophylaxis" refers to the regimen of preventing the onset of a disease or disorder such that the clinical symptoms of the disease or disorder do not develop. Thus, "preventing" involves administering therapy to a subject before signs of disease can be detected in the subject. The subject may be an individual at risk of developing a disease or disorder, such as an individual having one or more risk factors known to be associated with the development or onset of a disease or disorder.
As used herein, the term "therapeutically effective amount" or "effective amount" refers to an amount effective to elicit a desired biological or medical response, including an amount of an agent that is sufficient to effect such treatment of a disease when administered to a subject to treat the disease. The effective amount will vary depending on the particular agent and the characteristics of the subject to be treated, such as age, weight, etc. An effective amount may include a range of amounts. As understood in the art, an effective amount may be one or more doses, i.e., a single dose or multiple doses may be required to achieve a desired therapeutic endpoint. An effective amount may be considered in the context of administration of one or more therapeutic agents, and if combined with one or more other agents, a single agent may be considered to be administered in an effective amount to achieve or achieve a desired or beneficial result. The appropriate dosage of any co-administered agents may optionally be reduced by the combined effects (e.g., additive or synergistic effects) of the agents.
As used herein, "co-administration" includes administration of a unit dose of an agent disclosed herein before or after administration of a unit dose of one or more additional therapeutic agents, e.g., administration of an agent disclosed herein within seconds, minutes, or hours of administration of one or more additional therapeutic agents. For example, in some embodiments, a unit dose of an agent of the present disclosure is administered first, followed by a unit dose of one or more additional therapeutic agents within seconds or minutes. Alternatively, in other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by a unit dose of a compound of the present disclosure within seconds or minutes. In some embodiments, a unit dose of a compound of the present disclosure is administered first, followed by administration of the unit dose of one or more additional therapeutic agents after several hours (e.g., 1 hour-12 hours). In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound of the disclosure after several hours (e.g., 1 hour-12 hours).
Administration "in combination" with "one or more additional therapeutic agents includes simultaneous (concurrent) and sequential or sequential administration in any order.
The term "simultaneously" is used herein to refer to the administration of two or more therapeutic agents, wherein at least portions of the administrations overlap in time or wherein the administration of one therapeutic agent falls within a short period of time relative to the administration of the other therapeutic agent. For example, two or more therapeutic agents are administered at intervals of no more than about a specified number of minutes.
The term "sequentially" is used herein to refer to the administration of two or more therapeutic agents, wherein the administration of one or more agents continues after the administration of one or more other agents is discontinued, or wherein the administration of one or more agents begins before the administration of one or more other agents. For example, administration of two or more therapeutic agents is administered at intervals exceeding about a specified number of minutes.
As used herein, "combined" refers to administration of one form of treatment in addition to another form of treatment. Thus, "binding" refers to the administration of one form of treatment before, during, or after the administration of another form of treatment to an individual.
The term "conjugate" or "antibody-drug conjugate" refers to an antibody that is chemically linked to a second chemical moiety, such as a therapeutic agent or a cytotoxic agent. The term "pharmaceutical agent" includes chemical compounds, mixtures of chemical compounds, biological macromolecules, or extracts made from biological materials. In some embodiments, therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, paclitaxel, cytochalasin B, poncirin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide (teniposide), vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthrax, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin, and analogs or homologs thereof. When used in the context of an immunoassay, the conjugate antibody may be a detectably labeled antibody that serves as a detection antibody.
"intravenous administration" is the administration of a substance directly into a vein or "intravenous". The Intravenous (IV) route is a faster way of delivering fluids and drugs to the whole body than other routes of administration. Infusion pumps may allow for precise control over the flow rate and total amount of drug delivered. However, in cases where the flow rate variation does not have serious consequences, or if the pump is not available, the drip is typically caused to flow simply by placing the bag above the patient's level and using a clamp to adjust the rate. Alternatively, if the patient requires a high flow rate and the IV access device has a diameter large enough to accommodate it, a rapid infuser may be used. This is an inflatable cuff placed around the fluid bag to force fluid into the patient or similar electrical device that can also heat the infused fluid. Intermittent infusion is used when the patient only needs medication at certain times, which does not require additional fluid. It may use the same technique as intravenous infusion (pump or gravity infusion), but disconnect the tubing from the IV access device after the full dose of drug has been administered. Some drugs are also administered by IV push or bolus, meaning that the syringe is connected to the IV access device and the drug is directly infused (slow infusion if the drug may irritate the vein or cause too fast an effect). Once the drug has been injected into the fluid flow of the IV tubing, some means is necessary to ensure that it reaches the patient from the tubing. Typically, this is accomplished by allowing the fluid stream to flow normally and thereby carry the drug into the blood stream; however, sometimes a second fluid injection is used after injection as a "flush" to push the drug into the blood stream faster. Thus, in one embodiment, the agent or combination of agents described herein can be administered in the following manner: administration is by IV alone or in combination with administration of certain components of the treatment regimen by oral or parenteral routes.
"oral administration" is a route of administration in which the substance is taken up through the oral cavity and includes buccal, sub-labial and sublingual administration as well as parenteral administration, and through the respiratory tract unless done by, for example, a tube, such that the drug does not come into direct contact with any oral mucosa. Typical forms of oral administration of therapeutic agents include the use of tablets or capsules. Thus, in one embodiment, a compound or combination of compounds described herein can be administered in the following manner: by the oral route alone or in combination with administration of certain components of the treatment regimen by the IV or parenteral route.
Also provided herein are pharmaceutically acceptable salts, hydrates, solvates, tautomeric forms, polymorphs, and prodrugs of the compounds of formula (I) described herein. By "pharmaceutically acceptable" or "physiologically acceptable" is meant compounds, salts, compositions, dosage forms, and other materials suitable for human pharmaceutical use.
The compounds of formula (I) described herein may be prepared and/or formulated as pharmaceutically acceptable salts. Pharmaceutically acceptable salts are non-toxic salts of the free base form of the compound which have the desired pharmacological activity of the free base. These salts may be derived from inorganic acids, organic acids or bases. For example, a compound containing basic nitrogen may be prepared as a pharmaceutically acceptable salt by contacting the compound with an inorganic or organic acid. Non-limiting examples of pharmaceutically acceptable salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, octanoate, acrylate, formate, isobutyrate, hexanoate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1, 4-dioate, hexyne-1, 6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, sulfonate, methylsulfonate, propylsulfonate, benzenesulfonate, xylenesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, γ -hydroxybutyrate, glycolate, tartrate and mandelate. A list of other suitable pharmaceutically acceptable salts can be found in Remington, the Science and Practice of Pharmacy, 21 st edition, lippincott Wiliams and Wilkins, philiadelphia, pa., 2006.
Non-limiting examples of "pharmaceutically acceptable salts" of the compounds of formula (I) disclosed herein also include those derived from suitable bases such as alkali metals (e.g., sodium, potassium), alkaline earth metals (e.g., magnesium), ammonium and NX 4 + (wherein X is C 1 -C 4 Alkyl). Also included are base addition salts such as sodium or potassium salts.
MCL-1 inhibitors
Compounds of formula (I)
In some embodiments, provided herein is a method of treating cancer, the method comprising: administering to a human patient in need thereof a therapeutically effective amount of an antibody-drug conjugate and a therapeutically effective amount of an MCL-1 inhibitor;
wherein the antibody-drug conjugate comprises an anti-Trop-2 antibody and an anti-cancer agent; and is also provided with
Wherein the MCL-1 inhibitor has formula (I) or is a pharmaceutically acceptable salt thereof:
wherein R is 1 Is a 5-10 membered heteroaryl group containing 1-2 heteroatoms; wherein each heteroatom is independently selected from nitrogen, sulfur, and oxygen;
R 1 optionally independently selected from halogen, hydroxy, -CN, C 1-6 Alkyl, C 1-6 Haloalkyl, -OR 7 And C 3-6 1-3 substituents of cycloalkyl; and is also provided with
Each R 2 、R 3 、R 4 And R is 5 Independently hydrogen or C 1-6 An alkyl group;
R 6 hydrogen or halogen; and is also provided with
R 7 Independently hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl and C 3-10 Cycloalkyl groups.
In some embodiments of the methods described herein, the MCL-1 inhibitor is a compound of formula (II):
Each R 1 、R 2 、R 3 、R 4 、R 5 And R is 6 As defined above or elsewhere in this disclosure.
In some embodiments, the MCL-1 inhibitor is a compound of formula (III),
or a pharmaceutically acceptable salt thereof:
in some embodiments, the MCL-1 inhibitor is a compound of formula (I), formula (II) or formula (III), or a pharmaceutically acceptable salt thereof, R 2 Is hydrogen. In some embodiments, R 2 Is C 1-3 An alkyl group. R is R 2 Is methyl.
In some embodiments, the MCL-1 inhibitor is of formula (I)(I) A compound of formula (II) or formula (III) or a pharmaceutically acceptable salt thereof, R 3 Is C 1-3 An alkyl group. In some embodiments, R 3 Is methyl.
In some embodiments, the MCL-1 inhibitor is a compound of formula (I), formula (II) or formula (III), or a pharmaceutically acceptable salt thereof, R 4 Is hydrogen. In some embodiments, R 5 Is C 1-3 An alkyl group. In some embodiments, R 5 Is methyl. In some embodiments, R 6 Is Cl.
In some embodiments, the MCL-1 inhibitor is a compound of formula (I), formula (II) or formula (III), or a pharmaceutically acceptable salt thereof, R 1 To optionally be C 1-4 Alkyl and C 1-4 Alkoxy substitutedIn some embodiments, R 1 Is optionally-CH 3 and-OCH 3 Substituted->In some embodiments, R 1 is-CH 3 and-OCH 3 Substituted->In some embodiments, R 1 Is->
In some embodiments, the MCL-1 inhibitor is compound A (N- [ (4S, 7aR,9aR,10S,11E, 14S) -6 '-chloro-10-methoxy-14-methyl-16-oxo-18-oxo-3', 4', 7a,8, 9a,10,13,14,15, 18-dodecahydro-2' H-spiro [1,19- (binadimethy) -16 lambda ] 4 Cyclobutane [ i ]][1,4]Oxazepino [3,4-f][1,2,7]Thiadiazepine hexadecyne-4, 1' -naphthalene]-16-yl]-3-methoxy-1-methyl-1H-pyrazole-4-carboxamide) and has the following structure:
compound a is described in USPN 10,703,733 and example 154 of WO 2019/222112, which are incorporated herein by reference.
In some embodiments, the MCL-1 inhibitors that may be administered include, but are not limited to: compounds disclosed in USPN 10,703,733 (Gilead Sciences), AMG-397, AMG-176, PRT-1419, S64315, AZD59991, ABBV-467, WO2019222112 (gilide Sciences), WO2021096860 (gilide Sciences), WO2017147410 (ampen)), WO2019046150 (ampen), a WO2019046150 (Advance Co.), WO2019046150 (AstraZeneca), WO2019046150 (Ab Linekang), A. Al Linekang) WO2019046150 (AbbVie), WO2019046150 (Prelude), WO2019046150 (AbbVie) WO2019046150 (Ebervy), WO2019046150 (Ebervy) WO2019046150 (Abovi Co., ltd.), WO2019046150 (Abovi Co., ltd.) WO2019046150 (Abovi Co., ltd.), WO2019046150 (Abovi Co., ltd.), compounds disclosed in WO2020078875 (Shi Weiya), WO2017125224 (Shi Weiya), WO2020236817 (Shi Weiya), WO2016207226 (Shi Weiya), WO2016207217 (Shi Weiya), WO2016207216 (Shi Weiya) and WO2007147613 (Novartis).
In some embodiments, the MCL-1 inhibitor is selected from the group consisting of AMG-397, AMG-176, PRT-1419, and S64315. In some embodiments, the MCL-1 inhibitor is AMG-176. In some embodiments, the MCL-1 inhibitor is AMG-397. In some embodiments, the MCL-1 inhibitor is PRT-1419. In some embodiments, the MCL-1 inhibitor is S64315.
The compounds disclosed herein may contain one or more asymmetric centers and thus may produce enantiomers, diastereomers, and other stereoisomeric forms that may be defined as (R) -or (S) -depending on the absolute stereochemistry. The present disclosure is intended to include all such possible isomers and their racemic and optically pure forms. Optically active (+) and (-) or (R) -and (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques (e.g., chromatography and fractional crystallization). Conventional techniques for preparing/separating the individual enantiomers include chiral synthesis from suitable optically pure precursors or resolution of the racemate (or of a salt or derivative) using, for example, chiral High Pressure Liquid Chromatography (HPLC). Also, all tautomeric forms are intended to be included.
Formulations
In the methods provided herein, the MCL-1 inhibitor may be administered as a pharmaceutical composition. In certain embodiments, the pharmaceutical composition comprises a compound of formula (I), (II), (III) or compound a or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In certain embodiments, the pharmaceutical composition comprises one or more additional therapeutic agents, as described in more detail below.
Pharmaceutical compositions comprising the MCL-1 inhibitors disclosed herein, or pharmaceutically acceptable salts thereof, may be prepared with one or more pharmaceutically acceptable excipients, which may be selected according to conventional practice. "pharmaceutically acceptable excipients" include, but are not limited to, any auxiliary, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonizing agent, solvent, or emulsifier that has been approved by the U.S. food and drug administration as acceptable for use in humans or livestock.
In certain embodiments, the pharmaceutical composition is provided in a solid dosage form, a packIncluding solid oral dosage forms such as tablets. The tablets may contain excipients including glidants, fillers, binders, and the like. The aqueous compositions may be prepared in sterile form and, when intended for delivery by means other than oral administration, may generally be isotonic. All compositions may optionally contain excipients, such as those described in Rowe et al Handbook of Pharmaceutical Excipients,6 th edition, american Pharmacists Association,2009 (handbook of pharmaceutical excipients,6 th edition, american society of pharmacies, 2009). Excipients may include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrins, hydroxyalkyl celluloses, hydroxyalkyl methylcellulose, stearic acid, and the like.
The pharmaceutical compositions disclosed herein include those suitable for various routes of administration, including oral administration. The composition may be present in unit dosage form and may be prepared by any of the methods well known in the pharmaceutical arts. Such a method comprises the steps of: a step of associating an active ingredient (e.g., a compound of the present disclosure or a pharmaceutically acceptable salt thereof) with one or more pharmaceutically acceptable excipients. The compositions may be prepared by uniformly and intimately bringing into association the active ingredient with liquid excipients or finely divided solid excipients or both, and then, if necessary, shaping the product. Techniques and formulations can be found generally in Remington, the Science and Practice of Pharmacy, 21 st edition, lippincott Wiliams and Wilkins, philiadelphia, pa.,2006.
Compositions suitable for oral administration as described herein may be presented as discrete units (unit dosage forms) including, but not limited to, capsules, sachets or tablets each containing a predetermined amount of the active ingredient. In one embodiment, the pharmaceutical composition is a tablet.
In some embodiments, the tablet comprises compound a at an intensity of 5mg and 25 mg. In some embodiments, the tablet contains copovidone, lactose monohydrate, microcrystalline cellulose, polyvinylpyrrolidone, magnesium stearate, polyvinyl alcohol, titanium dioxide, polyethylene glycol, and talc.
Antibody-drug conjugates (ADC)
In some embodiments, the methods of treating cancer disclosed herein comprise: administering to a human patient in need thereof a therapeutically effective amount of an antibody-drug conjugate and a therapeutically effective amount of an MCL-1 inhibitor. In some embodiments, the antibody-drug conjugate comprises an anti-Trop-2 antibody and an anti-cancer agent.
In some embodiments, the antibody-drug conjugate is salmeterol xinafoate gordonin disclosed in USPN 7,999,083. In some embodiments, the ADC comprises an antibody-drug conjugate disclosed in USPN 7,999,083, which is incorporated herein by reference. In some embodiments, sha Xituo bead mab goretinide is Sha Xituo bead mab goretinide-hziy.
In some embodiments, the anti-Trop-2 antibody-drug conjugate is dapobutan Shan Kangde lutecan (datopotamab deruxtecan). In some embodiments, the administrable anti-Trop-2 antibody-drug conjugates include, but are not limited to, the conjugates disclosed in USPN 9,850,312, WO 20240467 and WO 18036438.
In some embodiments, the antibody portion of the ADC is an IgG antibody or antigen-binding antibody fragment. Antibodies may be of various isotypes, preferably human IgG 1, 55IgG2, igG3 or IgG4, more preferably comprising human IgG 1 hinge and constant region sequences. The antibody or fragment thereof may be a chimeric human-mouse, chimeric human-primate, humanized (human framework and murine hypervariable (CDR) regions) or fully human anti-60 antibody, and variants thereof such as a half IgG4 antibody (known as "monomer"), as described by van der Neut Kolfschoten et al (Science 2007; 317:1554-1557). More preferably, the antibody or fragment thereof may be designed or selected to comprise a human constant region 65 sequence belonging to a particular allotype, which may result in reduced immunogenicity when the antibody or ADC is administered to a human subject. Preferred allotypes for administration include non-Glml allotypes (nlml), such as Glm3, glm3,1, glm3,2 or Glm3,1,2. More preferably, the allotype is selected from the group consisting of: nGlml, glm3, nGlml,2 and Km3 allotypes.
In some embodiments, the antibody portion of the ADC is an anti-Trop-2 antibody. In some embodiments, anti-Trop-2 antibodies include, but are not limited to: TROP2-XPAT (Amunix), BAT-8003 (BioTheta Biotechnology Co., ltd. (Shanghai Junshi Biosciences)), TROP 2-IR700 (Chiome Bioscience), daptomtan Shan Kangde Lutekang (Daiichi Sankyo, aoslikang Co., ltd.), GQ-1003 (Kide medicine Co., genequantum Healthcare, sanxinum biological preparation Co., ltd. (Samsung BioLogics)), DAC 002 (Hangzhou Multi-happy Biotechnology Co., ltd. (Hangzhou DAC Biotech), shanghai Junyi Biotechnology Co., ltd. (Shanghai Junshi Biosciences)), sha Xituo bead monoclonal antibody govintekang (Jili De science Co., ltd.), E1-3s (Immunomerics/Jileyd Co., IBC Pharmaceuticals) TROP2-TRACTr (Janux Therapeutics), LIV-2008 (LivTech/Chiome, yandol Corp., yakult Honsha), shanghai Fu Han Kummer Sida Biotechnology Co., ltd (Shanghai Henlius BioTech)), LIV-2008b (LivTech/Chiome), anti-TROP-2 a (Oncooxx), anti-TROP-2 b (Oncooxx), OXG-64 (Oncooxx), OXS-55 (Oncooxx), humanized anti-TROP 2-SN38 antibody conjugate (Shanghai Katsuztec Co., ltd., TOT Biopharma), anti-TROP 2 antibody-CLB-SN-38 conjugate (Shanghai Fudan Zhangjia biological medicine Co., ltd.), shanghai Fudan-Zhangjia Bio-Pharmaceutical), SKB-264 (company (Sichuan Kelun Pharmaceutical) of the company of the medicine of the sciences of the Sichuan province)/United states of America (Klus Pharma)), TROP2-Ab8 (Abmart) of the company of the Aibi, TROP2-IgG (NMU) of the university of the Beijing medical science, 90Y-DTPA-AF650 (first Hospital of the university of Beijing), hRS7-CM (SynAffix), 89Zr-DFO-AF650 (University of Wisconsin-Madison) of the university of the Wis, anti-TROP 2 antibodies (Mediterranea Theranostic, legoChem Biosciences) and KD-065 (Nanjing KAEDI Biotech) of the biological sciences of the North Beijing).
Additional examples of anti-TROP-2 therapeutic agents include, but are not limited to: E1.BB.3z-92MI (immunomedia/Gillede Corp.), anti-Trop-2 CAR-T (Gillede Corp.), trop-2CAR-T (Hangzhou Lonzyme Biological Technology) of Hangzhou Rong Ze Biotechnology Co., ltd.), ARB-001 (Arbele) and MT-103 (Myeloid Therapeutics).
Examples of anti-TROP-2 antibodies include, but are not limited to: WO2020016662 (Aibi Co., ltd.), WO2020249063 (Baiyatai Biopharmaceutical Co., ltd.), US20190048095 (Baiyatai Biopharmaceutical Co., ltd.), WO2013077458 (LivTech/Chiome), EP 2013077458 (Chiome), WO2013077458 (first Co., ltd.), WO2013077458 (first co-product ), US 2013077458 (first co-product, first co-product), CN 2013077458 (boozhen company (biossion)) US 2013077458 (immunomedia/Gillede Co.), WO2013077458 (immunomedia/Gillede Co.), US 2013077458 (immunomedia/Gillede Co.), A. When the composition is used, the composition is used in a medical treatment US 2013077458 (immunomedia/Gillede Co.), A. RTM. And B. US 2013077458 (immunomedia/Gillede Co.), WO2013077458 (immunomedia/Gillede Co.), US 2013077458 (immunomedia/Gillede Co.), CN 2013077458 (immunomedia/Gillede Co.), A. RTM. And B. US 2013077458 (immunomedia/Gillede Co.), WO2013077458 (immunomedia/Gillede Co.), a kit for treating a disorder US 2013077458 (immunomedia/Gillede Co.), CN 2013077458 (immunomedia/Gillede Co.), A/C, those described in WO2014163684 (Gilreden), US9427464 (LivTech/Chiome), US10501555 (Abruzzo Theranostic/Oncxx), WO2018036428 (Sichuan Korea pharmaceutical Co., ltd.), WO2013068946 (Pfizer)), WO2007095749 (Roche) and WO2020094670 (SynAffix).
Other examples of anti-TROP-2 therapeutic agents include, but are not limited to, those described in WO2016201300 (gilid corporation) and CN108440674 (the biotechnology company, rong Ze, hangzhou).
In some embodiments, the anti-Trop-2 antibody is selected from the group consisting of hRS7, trop-2-XPAT, and BAT-8003.
In some embodiments, the anti-Trop-2 antibody is hRS7. In some embodiments, hRS7 is as disclosed in U.S. patent nos. 7,238,785, 7,517,964, and 8,084,583, which are incorporated herein by reference. Additional publications of hRS7 include international patent publication WO2003074566.
In some embodiments, the antibody-drug conjugate comprises an anti-Trop-2 antibody and an anti-cancer agent linked by a linker. In some embodiments, the linker comprises a linker disclosed in USPN 7,999,083. In some embodiments, the linker is CL2A.
In some embodiments, the drug moiety of the antibody-drug conjugate is a chemotherapeutic agent. In some embodiments, the chemotherapeutic agent is selected from the group consisting of Doxorubicin (DOX), epirubicin, morpholino doxorubicin (morpholino-DOX), cyanomorpholino-doxorubicin (cyanomorpholino-DOX), 2-pyrrolinyl-doxorubicin (2-PDOX), CPT, 10-hydroxycamptothecin, SN-38, topotecan (topotecan), lurtotecan (lurotecan), 9-aminocamptothecin, 9-nitrocamptothecin, taxane, geldanamycin, ansamycin, and epothilone. In some embodiments, the chemotherapeutic moiety is SN-38.
Formulations
Suitable routes of administration for ADCs include, but are not limited to: oral, parenteral, subcutaneous, rectal, transmucosal, enteral administration, intramuscular, intramedullary, intrathecal, direct intraventricular, intravenous, intravitreal, intraperitoneal, intranasal or intraocular injection. Alternatively, one may administer the compound locally rather than systemically, e.g., via injection of the compound directly into a solid tumor.
The ADC may be formulated according to known methods to prepare pharmaceutically useful compositions whereby the ADC is combined in a mixture with pharmaceutically suitable excipients. The ADC may be formulated for intravenous administration via bolus injection, slow infusion, or continuous infusion, for example. In some embodiments, the antibody is infused over a period of time less than about 4 hours. In some embodiments, the antibody is infused over a period of time less than about 3 hours. For example, the first 25mg to 50mg may be infused within 30 minutes or 15 minutes, and the remaining dose infused over the next 2 hours to 3 hours. The injectable formulations may be presented in unit dosage form, for example, in ampoules or in multi-dose containers, with the addition of preservatives. The composition may take the form of a suspension, solution or emulsion, such as in an oily or aqueous vehicle, and may contain a formulation, such as a suspending, stabilizing and/or dispersing agent. Alternatively, the active ingredient may be in powder form for use after dissolution with a suitable vehicle (e.g., sterile pyrogen-free water).
Therapeutic method
In some embodiments, the present disclosure provides a combination of an MCL-1 inhibitor and an antibody-drug conjugate for use in the treatment of cancer. In some embodiments, the antibody-drug conjugate is cetuximab, goretinide, and the MCL-1 inhibitor is compound a.
In some embodiments, the cancer is a Trop-2 expressing cancer.
In some embodiments, the cancer is selected from breast cancer, cervical cancer, colorectal cancer, endometrial cancer, epithelial ovarian cancer, esophageal cancer, follicular thyroid cancer, gastric or gastroesophageal junction adenocarcinoma, head and neck cancer, lung cancer, hepatocellular carcinoma, non-small cell lung cancer, ovarian cancer, prostate cancer, renal cell carcinoma, small cell lung cancer, urothelial cancer, and urinary system cancer.
In some embodiments, the cancer is selected from the group consisting of Triple Negative Breast Cancer (TNBC), hr+/HER 2-breast cancer, urothelial cancer, non-squamous non-small cell lung cancer (NSCLC), small Cell Lung Cancer (SCLC), head and neck squamous cell malignancy (HNSCC), and Muscle Invasive Bladder Cancer (MIBC).
In some embodiments, the cancer is metastatic. In some embodiments, the cancer is refractory.
In some embodiments, the cancer is selected from metastatic non-squamous non-small cell lung cancer (mNSCLC), metastatic triple negative breast cancer (mTNBC), and metastatic soft tissue malignant tumor with a non-specific tissue structure.
In some embodiments, the cancer is metastatic non-squamous non-small cell lung cancer (mNSCLC). In some embodiments, the cancer is metastatic triple negative breast cancer (mTNBC). In some embodiments, the cancer is a metastatic soft tissue malignancy with non-specific tissue architecture.
In some embodiments, the human patient has received at least one additional therapy prior to treatment with the combination therapy of the MCL-1 inhibitor and the antibody-drug conjugate. In some embodiments, the human patient has failed other therapies prior to the treatment disclosed herein. In some embodiments, the human patient has failed a chemotherapy.
In some embodiments, the human patient has failed therapy with an anti-PD 1 or anti-PDL 1 agent prior to treatment with the combination therapy of the MCL-1 inhibitor and the antibody-drug conjugate.
In some embodiments, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered simultaneously or separately with the antibody-drug conjugate.
In some embodiments, the MCL-1 inhibitor is Compound A. Generally, the dosage of compound a administered to a human will vary depending on factors such as the age, weight, height, sex, general medical condition, and past medical history of the patient. It may be desirable to provide the recipient with a dosage of antibody-conjugate in the range of about 1mg/kg to 24mg/kg in a single intravenous infusion, although lower or higher dosages may also be administered as the case may be. The dosage for 70kg of patients is, for example, from 70mg to 1,400mg at 1mg/kg to 20 mg/kg. The dose may be repeated as desired, for example, once a week for 4 to 10 weeks, once a week for 8 weeks, or once a week for 4 weeks. The dose may also be administered less frequently, such as every other week for several months, depending on the need for maintenance therapy; or monthly or quarterly for several months. In some embodiments, the dosage includes, but is not limited to: 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg, 10mg/kg, 11mg/kg, 12mg/kg, 13mg/kg, 14mg/kg, 15mg/kg, 16mg/kg, 17mg/kg, 18mg/kg, 19mg/kg, 20mg/kg, 22mg/kg, 24mg/kg, 26mg/kg, 28mg/kg, 30mg/kg, 35mg/kg, 40mg/kg, 45mg/kg, 50mg/kg, 55mg/kg, 65mg/kg, 70mg/kg, 80mg/kg, 90mg/kg, 100mg/kg, 120mg/kg, 140mg/kg, 150mg/kg, 160mg/kg, 180mg/kg, 200mg/kg, 220mg/kg, 240mg/kg, 250mg/kg, 260mg/kg, 280mg/kg, 300mg/kg, 350mg, 400mg/kg, 600mg/kg, 700mg/kg, 600mg/kg, and 750 mg/kg. Any amount in the range of 1mg/kg to 300mg/kg may be used. Any amount in the range of 1mg/kg to 100mg/kg may be used.
In some embodiments, the dose is administered multiple times, once or twice a week. A minimum dose schedule of 4 weeks, 8 weeks, 16 weeks or longer may be used. The administration schedule may include administration once or twice a week, with an administration period selected from the group consisting of: (i) weekly; (ii) every other week; (iii) One week of treatment followed by two, three or four weeks of discontinuation; (iv) Treatment for two weeks followed by one week, two weeks, three weeks or four weeks of discontinuation; (v) Three weeks of treatment followed by one, two, three, four or five weeks of discontinuation; (vi) Four weeks of treatment followed by one, two, three, four or five weeks of discontinuation; (vii) Five weeks of treatment followed by one, two, three, four or five weeks of discontinuation; and (viii) monthly. The cycle may be repeated 4, 6, 8, 10, 12, 16, or 20 or more times.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered orally.
In some embodiments, the amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered at a dose of about 5mg/kg, 15mg/kg, or 50 mg/kg.
In some embodiments, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered at a dose of about 5 mg/kg.
In some embodiments, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered at a 21 day period, wherein administration is discontinued for 5 days after 2 days.
In some embodiments, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered for up to 105 weeks on days 1, 2, 8, 9, 15, and 16 of each 21 day cycle.
In some embodiments, the antibody-drug conjugate is administered in the form of intravenous infusion.
Generally, the dosage of antibody-drug conjugate administered to a human will vary depending on factors such as the age, weight, height, sex, general medical condition, and past medical history of the patient. It may be desirable to provide the recipient with a dosage of antibody-conjugate in the range of about 1mg/kg to 24mg/kg in a single intravenous infusion, although lower or higher dosages may also be administered as the case may be. The dosage for 70kg of patients is, for example, from 70mg to 1,400mg at 1mg/kg to 20 mg/kg. The dose may be repeatedly administered as desired, for example, once a week for 4 to 10 weeks, once a week for 8 weeks, or once a week for 4 weeks. The dose may also be administered less frequently, such as every other week for several months, depending on the need for maintenance therapy; or monthly or quarterly for several months. In some embodiments, the dosage includes, but is not limited to: 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg, 10mg/kg, 11mg/kg, 12mg/kg, 13mg/kg, 14mg/kg, 15mg/kg, 16mg/kg, 17mg/kg, 18mg/kg, 19mg/kg, 20mg/kg, 22mg/kg and 24mg/kg. Any amount in the range of 1mg/kg to 24mg/kg may be used. In some embodiments, the dose is administered multiple times, once or twice a week. A minimum dose schedule of 4 weeks, 8 weeks, 16 weeks or longer may be used. The administration schedule may include administration once or twice a week, with an administration period selected from the group consisting of: (i) weekly; (ii) every other week; (iii) One week of treatment followed by two, three or four weeks of discontinuation; (iv) Treatment for two weeks followed by one week, two weeks, three weeks or four weeks of discontinuation; (v) Three weeks of treatment followed by one, two, three, four or five weeks of discontinuation; (vi) Four weeks of treatment followed by one, two, three, four or five weeks of discontinuation; (vii) Five weeks of treatment followed by one, two, three, four or five weeks of discontinuation; and (viii) monthly. The cycle may be repeated 4, 6, 8, 10, 12, 16, or 20 or more times.
In some embodiments, the antibody-drug conjugate may be administered at one dose every 2 weeks or every 3 weeks, repeated for a total of at least 3 doses. Or twice weekly for 4 to 6 weeks. If the dosage is reduced to about 200 to 300mg/m 2 (340 mg per dose for 1.7m patient, or for)70kg patient 4.9 mg/kg), it may be administered once or even twice a week for 4 to 10 weeks. In some embodiments, the dosage schedule may be reduced, i.e., every 2 weeks or every 3 weeks, for 2 months to 3 months. However, it has been determined that for repeated dosing cycles, even higher doses (such as once a week or once every 2 weeks to every 3 weeks, 2mg/kg each) can be administered by slow intravenous infusion. With appropriate adjustments to the dosage and schedule, the dosing schedule may optionally be repeated at other intervals, and the dosages may be administered by various parenteral routes.
In some embodiments, the antibody-drug conjugate dose is administered on days 1 and 8 of each 21-day cycle.
In some embodiments, the antibody-drug conjugate is administered at a dose of between about 4mg/kg and about 12 mg/kg. In some embodiments, the antibody-drug conjugate is administered at a dose of between about 8mg/kg and about 12 mg/kg.
In some embodiments, the antibody-drug conjugate is administered at a dose of about 8mg/kg, about 10mg/kg, or about 12 mg/kg.
In some embodiments, the antibody-conjugate is cetuximab, goveritecan. In some embodiments, the dose of Sha Xituo beads of the anti-gaultherian is in the range of about 2mg/kg to 20mg/kg as a single intravenous infusion. In some embodiments, the dose of Sha Xituo beads of the anti-gaultherian is in the range of about 6mg/kg to 10mg/kg as a single intravenous infusion. In some embodiments, the dose comprises 2mg/kg, 2.5mg/kg, 3mg/kg, 3.5mg/kg, 4mg/kg, 4.5mg/kg, 5mg/kg, 5.5mg/kg, 6mg/kg, 6.5mg/kg, 7mg/kg, 7.5mg/kg, 8mg/kg, 8.5mg/kg, 9mg/kg, 9.5mg/kg, 10mg/kg, 10.5mg/kg, 11mg/kg, 11.5mg/kg, 12mg/kg, 12.5mg/kg, 13mg/kg, 13.5mg/kg, 14mg/kg, 14.5mg/kg, 15mg/kg, 15.5mg/kg, 16mg/kg, 16.5mg/kg, 17mg/kg, 17.5mg/kg, 18mg/kg, 18.5mg/kg, 19mg/kg, and 20.5 mg/kg. In some embodiments, the dosage of antibody-conjugate is 7.5mg/kg.
In some embodiments, the method further comprises one or more additional therapeutic forms selected from the group consisting of antibodies, conjugates, gene therapy, chemotherapy, radiation therapy, surgical therapy, BTK inhibitors, and checkpoint inhibitors.
In some embodiments, the method further comprises radiation therapy.
In some embodiments, the method further comprises administering one or more additional therapeutic agents. In some embodiments, the additional therapeutic agent is selected from the group consisting of a chemotherapeutic agent, a checkpoint inhibitor, a FLT3 agonist, and a BTK inhibitor.
In some embodiments, the checkpoint inhibitor is selected from the group consisting of an anti-PD-1 agent, an anti-PD-L1 agent, an anti-PD-1/PD-L1 interaction inhibitor, an anti-CTLA 4 agent, and an anti-TIGIT agent.
In some embodiments, the FLT3 agonist is GS-3583.FLT3 agonists also include CDX-301 and agents disclosed in PCT publication WO2020/263830A 1.
In some embodiments, the FLT3 agonist is an Fc fusion protein disclosed in WO 2022031876.
In some embodiments, the present disclosure provides a method for treating cancer. The method comprises administering an MCL-1 inhibitor and an antibody-drug conjugate to treat cancer; and the method further comprises administering one or more additional therapeutic agents, provided that the additional therapeutic agent is not a FLT3 agonist. In some embodiments, the additional therapeutic agent is not a FLT3-Fc fusion protein. In some embodiments, the antibody-drug conjugate is cetuximab, goretinide, and the MCL-1 inhibitor is compound a; and the additional therapeutic agent is not a FLT3 agonist. In some embodiments, the additional therapeutic agent is not a FLT3 agonist as disclosed in WO 2020/263830. In some embodiments, the additional therapeutic agent is not a fusion protein comprising the amino acid sequence of SEQ ID NO. 14 of U.S. patent No. 11/124,582.
In some embodiments, the method comprises administering a checkpoint inhibitor. In some embodiments, the checkpoint inhibitor is selected from the group consisting of an anti-PD-1 agent, an anti-PD-L1 agent, an anti-PD-1/PD-L1 interaction inhibitor, an anti-CTLA 4 agent, and an anti-TIGIT agent. In some embodiments, the checkpoint inhibitor is selected from the group consisting of nivolumab, pembrolizumab, atenolizumab, sirolimus, and pilizumab. In some embodiments, the checkpoint inhibitor is selected from the group consisting of ipilimumab (ipilimumab), lambrolizumab, tremelimumab (tremelimumab), dulcitol You Shan antibody (durvalumab), avermectin (avelumab), domvanalimab, and tireli Li Youshan antibody (tiragolumab).
Examples of CTLA4 inhibitors that can be co-administered include, but are not limited to: ipilimumab, tremelimumab, BMS-986218, AGEN1181, AGEN1884, BMS-986249, MK-1308, REGN-4659, ADU-1604, CS-1002, BCD-145, APL-509, JS-007, BA-3071, ONC-392, AGEN-2041, JHL-1155, KN-044, CG-0161, ATOR-1144, PBI-5D3H5, BPI-002, and multispecific inhibitors FPT-155 (CTLA 4/PD-L1/CD 28), PF-06936308 (PD-1/CTLA 4), MGD-019 (PD-1/CTLA 4), KN-046 (PD-1/CTLA 4), MEDI-5752 (CTLA 4/PD-1), xmAb-20717 (PD-1/CTLA 4) and AK 104 (CTLA 4/PD-1).
Examples of inhibitors of PD-L1 (CD 274) or PD-1 (PDCD 1) that may be co-administered include, but are not limited to, pembrolizumab, nivolumab, sibirinotecan, pirimab, AMP-224, MEDI0680 (AMP-514), sdazumab, abilizumab, avelumab, duvalli You Shan, ALN-PDL, BMS-936559, CK-301, PF-06801591, BGB-108, BGB-A317 (tirelizumab), GLS-010 (WBP-3055), AK-103 (HX-008), GB-226, AK-105, CS-1003, HLX-10, MGA-012, BI-754091, PDR-001, AGEN-2034, and the like JS-001 (terlipressin Li Shan), JNJ-63723283, jenomab (CBT-501), LZM-009, BCD-100, LY-3300054, SHR-1201, SHR-1210 (Carrilizumab), sym-021, ABBV-181, PD1-PIK, BAT-1306, RO-6084 (PD-L1 antisense oligonucleotide), STI-1110, GX-P2, RG-7446, mDX-400, (MSB 0010718C), CX-072, CBT-502, TSR-042 (Duotalizumab), MSB-2311, JTX-4014, BGB-A333, SHR-1316, CS-1001 (KNP-3155), MEDI-0680, en Wo Lishan (envanfolib) (KN-035), KD-033, KY-1003, IBI-308 (Xindi Li Shan antibody), HLX-20, KL-A167, STI-A1014, STI-A1015 (IMC-001), BCD-135, FAZ-053, TQB-2450, MDX1105-01, MSB-0010718C, GS-4224, GS-4416, INCB086550, MAX10181, bispecific inhibitors FPT-155 (CTLA 4/PD-L1/CD 28), PF-06936308 (PD-1/CTLA 4), MGD-013 (PD-1/LAG-3), FS-118 (LAG-3/PD-L1) MGD-019 (PD-1/CTLA 4), KN-046 (PD-1/CTLA 4), MEDI-5752 (PD-1), RO-7121661 (PD-1/PD-3), xmAb-20717 (PD-1/CTLA 4), AK-104 (CTLA 4/PD-1), M-1/PD-L4 (CDL 1/PDL 1), PDL-35 (PDL-35, PDL-35 (PDL 1/PDL 1), PDL-35 (PDL-35) and PDL-35 (PDL-35) factor (PDL-35), PDL-35 (PDL-35) domain-35), and PDL-35 (PDL-35) protein (PDL-35) factor (PDL-35), and PDL-35 (CPL-35) protein (CPL 2), and PDL 2/CPL 2 domain protein (CPL 2/CPL 2), and PDL 2 protein (CPL 2/CPL 2).
Examples of PD-1 inhibitors include, but are not limited to, the compounds disclosed in the following patents: WO (Incote Corp), WO (Incote Co.), WO (Bezimuth Mitsui precious Co., ltd.) WO (Ensat Co., ltd.), WO (Ensat Co., ltd.) WO (inflieast corporation), WO (ziram corporation), WO (japanese guard pharmaceutical company Ltd) (Eisai Co Ltd); WO (), WO (; individual), WO (Incyte Corp), WO (Incyte Corp) WO (), WO (Incyte Corp), WO (), WO (), and WO () WO2016039749 (BristolMyers Squibb Co), WO2015019284 (Cambridge Enterprise Ltd), WO2016142894 (Aurigene Discovery Technologies Ltd), WO2015134605 (BristolMyers Squibb Co), WO2018051255 (Aurigene Discovery Technologies Ltd), WO2018051254 (Aurigene Discovery Technologies Ltd), WO2017222976 (Incyte Corp), WO2017070089 (Incyte Corp), WO2018044963 (BristolMyers Squibb Co), WO2013144704 (Aurigene Discovery Technologies Ltd), WO2018013789 (Incyte Corp), WO2017176608 (BristolMyers Squibb Co), WO2018009505 (BristolMyers Squibb Co), WO2011161699 (Aurigene Discovery Technologies Ltd), WO2015119944 (Incyte Corp; moxadong corporation (Merck Sharp & Dohme Corp)), WO2017192961 (inflieme corporation), WO2017106634 (inflieme corporation), WO2013132317 (Aurigene Discovery Technologies Ltd), WO2012168944 (Aurigene Discovery Technologies Ltd), WO2015036927 (Aurigene Discovery Technologies Ltd), WO2015044900 (Aurigene Discovery Technologies Ltd) and WO2018026971 (Arising International).
The PD-1/PD-L1 inhibitor may be administered in any suitable amount known to those skilled in the art. In some embodiments, the compound of formula I is administered to the subject in an amount of 0.1mg to 1000 mg. Representative amounts of PD-1/PD-L1 inhibitor administered to a subject include, but are not limited to, 0.1mg to 500mg, 1mg to 100mg, 1mg to 50mg, or 10mg to 50mg. Other amounts of PD-1/PD-L1 inhibitor administered to a subject include, but are not limited to, about 1mg or 2mg, 3mg, 4mg, 5mg, 6mg, 7mg, 8mg, 9mg, 10mg, 15mg, 20mg, 25mg, 30mg, 35mg, 40mg, 45mg, 50mg, 55mg, 60mg, 65mg, 70mg, 75mg, 80mg, 85mg, 90mg, 95mg, or about 100mg.
In some embodiments, the methods as described herein further comprise administering an anti-TIGIT antibody, such as BMS-986207, RG-6058, domvanalimab, AB, 57308, or AGEN-1307.
In some embodiments, the methods as described herein further comprise administering a BTK (bruton's tyrosine kinase) inhibitor. Examples of such BTK inhibitors are the compounds disclosed in us patent 7,405,295. Additional examples of BTK inhibitors include, but are not limited to: (S) -6-amino-9- (1- (but-2-ynyl) pyrrolidin-3-yl) -7- (4-phenoxyphenyl) -7H-purin-8 (9H) -one, acartinib (ACP-196), BGB-3111, HM71224, ibrutinib, M-2951, tiratinib (ONO-4059), PRN-1008, capetinib (CC-292) and TAK-020. In some embodiments, the BTK inhibitor is selected from the group consisting of acartinib, tiratinib, zebutinib, and PCI-32765.
In some embodiments, the method further comprises administering a chemotherapeutic agent. In some embodiments, the anticancer agent is selected from the group consisting of Doxorubicin (DOX), epirubicin, morpholino doxorubicin (morpholino-DOX), cyanomorpholino-doxorubicin (cyanomorpholino-DOX), 2-pyrrolinyl-doxorubicin (2-PDOX), CPT, 10-hydroxycamptothecin, SN-38, topotecan, lurtotecan, 9-aminocamptothecin, 9-nitrocamptothecin, taxane, geldanamycin, ansamycin, and epothilone. In some embodiments, the chemotherapeutic agent is docetaxel. In some embodiments, the chemotherapeutic agent is gemcitabine. In some embodiments, the chemotherapeutic agent is paclitaxel.
In certain embodiments, when the agents of the present disclosure are combined with one or more additional therapeutic agents as described herein, the components of the composition are administered in a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. In certain embodiments, when the agents of the present disclosure are combined with one or more additional therapeutic agents as described herein, the components of the composition are administered in a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. In certain embodiments, when the agents of the present disclosure are combined with one or more additional therapeutic agents as described herein, the components of the composition are administered in a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
Co-administration of an agent disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of the agent disclosed herein and one or more additional therapeutic agents such that a therapeutically effective amount of each agent is present in the patient.
Co-administration includes administering a unit dose of an agent disclosed herein before or after administering a unit dose of one or more additional therapeutic agents. The agents disclosed herein may be administered within seconds, minutes, or hours of administration of one or more additional therapeutic agents. For example, in some embodiments, a unit dose of an agent disclosed herein is administered first, followed by administration of the unit dose of one or more additional therapeutic agents within seconds or minutes. Alternatively, in other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of the unit dose of the agents disclosed herein within seconds or minutes. In some embodiments, a unit dose of an agent disclosed herein is administered first, followed by administration of the unit dose of one or more additional therapeutic agents after several hours (e.g., 1-12 hours). In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of the unit dose of the agents disclosed herein after several hours (e.g., 1-12 hours).
In some embodiments, the present disclosure provides a method of treating or preventing cancer. In certain embodiments, the present disclosure provides a method of treating or preventing cancer comprising administering to a patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the cancer is a hematological malignancy. In some embodiments, the cancer is multiple myeloma. In some embodiments, the cancer is selected from the group consisting of: breast cancer, colorectal cancer, skin cancer, melanoma, ovarian cancer, renal cancer, small cell lung cancer, non-small cell lung cancer, lymphoma, and leukemia. In some embodiments, the cancer is acute myelogenous leukemia.
In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is bladder cancer.
Examples
Example 1: in vitro synergy of MCL-1 inhibitors and SN-38 in TNBC and NSCLC cell lines
To test the combined potential between compound a and SN-38 (topoisomerase inhibitor), in vitro studies were performed in a panel of triple negative breast cancer (TNBC, n=3) and non-small cell lung cancer (NSCLC, n=2) cell lines using a synergistic Bliss independent model. Cells were exposed to a dose titration matrix of each compound alone and in combination for 72 hours, and Cell viability was then determined by Cell Titer Glo reagent. A strong Bliss synergy score (> 100) was observed across all cell lines tested.
Materials and methods
Cell culture and reagents
HCC70 #CRL-2315)、HCC1806(/>CRL-2335)、HCC1187(/>CRL-2322)、NCI-H522(/>CRL-5810) and H820 (-)>HTB-181) cell lines were thawed from liquid nitrogen storage and maintained in RPMI-1640 (Gibco-12633) +10% HI-FBI (Gibco-16140) +Pen/Strep (100X Gibco-15140) according to ATCC guidelines. Cells were passaged according to ATCC guidelines with 0.25% trypsin/EDTA (1X GIBCO-25200).
SN-38 and compound a stock (supplied by the gilled corporation sample library) were dispensed directly into the treatment wells to 0.1% v/v using a D300e digital dispenser (supplier) using DMSO (Sigma-D2438) as vehicle control.
According to the protocol for manufacturing microplates, cell Titer Glo was used TM (Promega) #G9241) to perform viability assessment and read luminescence on a Biotek Synergy Neo plate reader.
Cell viability combinatorial assays
For the synergy matrix assay, cell lines were seeded at 5,000 cells per well in 100 μl of recommended cell culture medium in a white transparent bottom 96-well plate (Corning # 3909). The treatment panels consisted of single agent dose responses to compound a (seven 3-fold dilutions plus untreated controls) or SN-38 (nine 3-fold dilutions plus untreated controls) and a checkerboard matrix of 63 different combinations. The concentration ranges are selected based on the relative sensitivity of each cell line to the compound. Five plates were used for each combination to generate enough replicates to calculate a synergy score with 95% confidence interval (95% CI).
Compounds and DMSO vehicle were applied to cells according to a checkerboard matrix using HP D300 dispenser directly aliquoted into the medium and at 37 ℃/5% co 2 Viability was measured by Cell Titer Glo after Wen Yoda hours at 100% relative humidity.
Data analysis
The excel template described by Prichard and Shipman { Prichard 1990} was used to evaluate the combined vitality data for synergy. Specifically, the individual component dose curves for SN-38 and compound a were normalized to percent viability on each plate and averaged across five technical replicates to calculate a theoretical additive killing of the combination according to the Bliss independent principle. The calculated values were compared with experimental results generated in a sixty-three concentration checkerboard. Depending on whether the observed growth inhibition is greater or less than the calculated value, a synergy or antagonism score is generated, respectively.
For example, if two compounds (B) and (C) at a given concentration each resulted in 60% inhibition, their theoretical cumulative inhibition would be 84% according to the following Bliss independent:
60% B +60% C *(100%-60% B )=84% B+C
if the experimental result is greater than calculated (e.g., 90% inhibition), the difference [6% ] will be added to the synergy score. If the result is lower (e.g., 78% inhibition), the difference [6% ] will be added to the antagonism score.
These differences are summed across the entire checkerboard (sixty-three holes) to give a signal with unit μm 2 % cumulative synergy and antagonism score to reflect the 2D surface of the dose response. The 95% confidence interval adjustment was applied to the synergy and antagonism scores and each sum was compared to a scale based on the initial method: scores greater than 50 were considered moderate synergy, and scores greater than 100 were considered strong synergy and likely displayed a combined effect in vivo { Prichard 1990}.
Data for the combinatorial assays are presented in three forms. Synergy scores at 95% confidence interval averaged from n=2 assays. An exemplary list and graphical percentage inhibition matrix for each cell line. An exemplary list and graphical synergy matrix at 95% confidence interval for each cell line.
To test the combined potential between compound a and SN-38 (topoisomerase inhibitor), in vitro studies were performed in a panel of TNBC (n=3) and NSCLC (n=2) cell lines using a synergistic Bliss independent model. Cells were exposed to a dose titration matrix of each compound alone and in combination for 72 hours, and Cell viability was then determined by Cell Titer Glo reagent. A strong Bliss synergy score (> 100) was observed across all cell lines tested.
TABLE 1 Compound A+SN-38 Bliss synergy scores at 95% confidence intervals
Table 2. Percent hcc70 inhibition results (n=5) 1 st parallel assay
Table 3% hcc70 inhibition results (n=5) 2 nd parallel assay
/>
Table 4. Percent hcc1806 inhibition results (n=5) 1 st parallel assay
Table 5% hcc1806 inhibition results (n=5) 2 nd parallel assay
Table 6% results of hcc1187 inhibition (n=5) 1 st parallel assay
Table 7% hcc1187 inhibition results (n=5) 2 nd parallel assay
Table 8% results of hcc1187 inhibition (n=5) 3 rd parallel assay
Table 9. Percent h522 inhibition results (n=5) 1 st parallel assay
Table 10. Percent h522 inhibition results (n=5) 2 nd parallel assay
Table 11. Percent inhibition results of h820 (n=5) 1 st parallel assay
Table 12. Percent inhibition results of h820 (n=5) 2 nd parallel assay
* The SN-38 dose response was shifted to a maximum concentration of 1.0 μm to capture a large potential range of synergistic activity
Example 2: in vitro synergy of MCL-1 inhibitors and paclitaxel in TNBC cells
Materials and methods
Cell culture and reagents
HCC70 #CRL-2315) and HCC1806 (++>CRL-2335) was thawed from the liquid nitrogen storage device and maintained in RPMI-1640 (Gibco-12633) +10% HI-FBI (Gibco-16140) +pen/Strep (100 x Gibco-15140) according to ATCC guidelines. MDA-MB-468 (+) >HTB-132) was thawed and maintained in DMEM (Gibco-11995) +10% HI-FBS+Pen/Strep. Cells were passaged according to ATCC guidelines with 0.25% trypsin/EDTA (1X GIBCO-25200).
Paclitaxel and compound a stock (supplied by the gilled corporation sample bank) were dispensed directly into the treatment wells to 0.1% v/v using a D300e digital dispenser (supplier) using DMSO (Sigma-D2438) as vehicle control.
According to the protocol for manufacturing microplates, cell Titer Glo was used TM (Promega #G9241) to perform viability assessment and read luminescence on a Synergy Neo2 reader.
Cell lysates for MSD assays were generated using 1 Xlysis buffer (10 Xcell signaling CST-9803), 100 Xprotease inhibitor, phosphatase inhibitor I, phosphatase inhibitor II (Meso Scale Discovery Inhibitor Pack R70AA-1 and PMSF (SIGMA catalog number 7626)
MCL-BAK and MCL1-BIM dimer assays, as well as the overall MCL1 assay, were developed by MSD Custom Assay Services and run using MSD U-PLEX Development Pack (K15227N) and revision of the protocol, "revision 2 of month 3 2018". GAPDH was determined by the MSD using a standard assay K151 PWD. All plates were read using MSD Read Buffer T (R92 TC) at MSD SECTOR Imager 2400.
Protein Simple reagent: EZ Standard Pack1 (PS-ST 01EZ: biotinyl)an ated ladder, FL standard and DTT), perox (044-379), luminal-S (043-311), antibody Dilution Buffer (042-203), strepitavidin HRP (042-414), secondary antibodies: sheep anti-rabbit (042-206) and sheep anti-mouse (042-205), separation Matrix (042-512), stacking Matrix (042-513), 10X Sample Buffer (042-195), wash Buffer (042-520), upper Running Buffer (043-163), lower Running Buffer (043-162), 384 well plate (040-663), size caps (55700), protein Simple Instruments Peggy Sue TM And Sally Sue TM . An antibody: MCL1 (CST-94296), FBXW7 (Abcam 109617 and Abcam 171961)
Cell viability combinatorial assays
For the synergy matrix assay, the TNBC cell line was seeded at 10,000 cells per well in 100 μl of the recommended cell culture medium in a white clear bottom 96-well plate (Corning # 3909). The plates were incubated at 37 ℃ and 100% rh for 20 hours, followed by compound exposure. The treatment profile consisted of a single dose response of the checkerboard matrix for compound a (seven 3-fold dilutions from 3 μm to 4nM plus untreated control) or paclitaxel (nine 3-fold dilutions from 3 μm to 0.5nM plus untreated control) and 63 different combinations. Five plates were used for each combination to generate enough replicates to calculate a synergy score with 95% confidence interval (95% CI).
Paclitaxel and DMSO vehicle were first applied to cells according to a checkerboard matrix using an HP D300 dispenser directly aliquoted into the medium. Paclitaxel was incubated for 4 hours, then washed out by media removal and 2x 200 μl wash with pre-warmed complete media, and finally replaced with 100 μl pre-warmed complete media. Cells were then exposed to compound a using a D300 dispenser with the same checkerboard matrix and Wen Yoda hours, then viability was measured by Cell Titer Glo.
MSD assay
For MSD assays, TNBC cell lines were seeded at 25,000 cells per well in recommended cell culture medium in white clear bottom 96-well plates (Corning # 3909). The plates were incubated at 37 ℃ and 100% rh for 20 hours, followed by compound exposure. The paclitaxel and DMSO vehicle were first applied to the cells using an HP D300 dispenser directly aliquoted into the medium. Paclitaxel was incubated for 4 hours, then washed out by media removal and 2×150 μl wash with pre-warmed complete media, and finally replaced with 100 μl pre-warmed complete media. After a further 20 hours, samples were harvested by pipetting the supernatant and 125 μl of 1x lysis buffer was added to each well. The plates were simply set on ice and transferred to a shake table at 4 ℃ for 20 minutes. The plates were placed on dry ice, flash frozen for 10 minutes, and then stored at-80 ℃ before being tested.
MCL1 and MCL1-BAK and MCL1-BIM dimer assays were run using materials and protocols provided by MSD Custom Assay Services based on its U-Plex technology. Plates were first prepared using a standard U-PLEX capture antibody coating protocol and then washed 3 times with 150uL MSD wash buffer. 25 μl of the sample or standard is directly added to the plate, the plate is sealed and incubated at room temperature for 1 hour with shaking. The plates were washed 3 times with 150 μl of wash buffer per well, and 50 μl of antibody detection solution was added to each well of the MSD plate, the plates were sealed and incubated at room temperature for 1 hour with shaking. The plates were again washed 3 times with 150 μl wash buffer per well. Mu.l of 2x read buffer was added to each well and the plates were read on MSD SECTOR Imager 2400.
GAPDH assay kit (MSD) was run according to the manufacturer's protocol using 25 μl of lysate per sample directly added to the plate. The plates were sealed, incubated at room temperature for 1 hour with shaking, and then washed 3 times with 150 μl wash buffer per well. Mu.l of antibody detection solution was added to each well, the plate was sealed, incubated at room temperature for 1 hour with shaking, and then washed 3 times with 150. Mu.l of wash buffer per well. Then 150 μl of 2x read buffer was added to each well and the plates were measured on MSD SECTOR Imager 2400 for Electrochemiluminescence (ECL).
Protein Simple(Simple Western)
Simple Western immunoassay was performed in capillaries. Samples and reagents were loaded into assay plates and placed in Protein Simple Instrument. Cell lysates are automatically loaded into capillaries and separated by size as they migrate through the stacking and separation matrix. The separated proteins are then immobilized to the capillary wall via proprietary photosensitizing capture chemistry. The target protein was identified using a primary antibody and immunodetected using HRP conjugated secondary antibody and chemiluminescent substrate. And detecting and quantifying the obtained chemiluminescent signal.
FBXW7 expression was measured in the cell line after a 4 hour dose of 1 μm paclitaxel followed by Western wash out using Simple and overnight incubation. Lysates were prepared and diluted to 0.5ug/ml in 1 Xlysis buffer. Simple Western platform was run on 384 well plates.
Markers, internal ladder and DTT were provided in lyophilized form by Simple Western. The reagents were resuspended as described in the protocol. 20uL of water was added to the markers. 40ul of water was added to the DTT, and 20ul of 10 Xsample buffer and 20ul of DTT were mixed and referred to as Z buffer. The markers were loaded in 1A. 5ul of lysate was added to a 1.7ml Eppendorf tube. 1.2ul of Z reagent was added to each sample. The sample was heated at 100C for 5 minutes, allowed to cool, and then spun in a microcentrifuge for 30 seconds. The sample was loaded into wells A2-12. An antibody was diluted 1:50 in antibody dilution buffer (6ul+295 ul dilution buffer). 20ul of each antibody was loaded in lanes 2-12. Up to 8 antibodies were directed against the different antibodies of each row. Actin diluted 1:300 was used as a loading control. In addition, secondary goat anti-rabbit or goat anti-mouse was loaded as needed. Plates were spun at room temperature for 2.6k 10 min and loaded into the instrument and allowed to run overnight. The target protein was identified using a primary antibody and immunodetected using HRP conjugated secondary antibody and chemiluminescent substrate. And detecting and quantifying the obtained chemiluminescent signal.
Data analysis
Synergistic effects on Bliss
The excel template described by Prichard and Shipman { Prichard 1990} was used to evaluate the combined vitality data for synergy. Specifically, individual component dose curves of paclitaxel and compound a were normalized to percent viability on each plate and averaged across 5 technical replicates to calculate a theoretical additive killing of the combination according to the Bliss independent principle. The calculated values were compared with experimental results generated in a sixty-three concentration checkerboard. Depending on whether the observed growth inhibition is greater or less than the calculated value, a synergy or antagonism score is generated, respectively.
For example, if two compounds (B) and (C) at a given concentration each resulted in 60% inhibition, their theoretical cumulative inhibition would be 84% according to the following Bliss independent:
60% B +60% C *(100%-60% B )=84% B+C
if the experimental result is greater than calculated (e.g., 90% inhibition), the difference [6% ] will be added to the synergy score. If the result is lower (e.g., 78% inhibition), the difference [6% ] will be added to the antagonism score.
These differences are summed across the entire checkerboard (sixty-three holes) to give a signal with unit μm 2 % cumulative synergy and antagonism score to reflect the 2D surface of the dose response. The 95% confidence interval adjustment was applied to the synergy and antagonism scores and each sum was compared to a scale based on the initial method: scores greater than 50 were considered moderate synergy, and scores greater than 100 were considered strong synergy and likely displayed a combined effect in vivo { Prichard 1990}.
Data for the combinatorial assays are presented in three forms. Synergy scores at 95% confidence interval averaged from n=2 assays. An exemplary list and graphical percentage inhibition matrix for each cell line. An exemplary list and graphical synergy matrix at 95% confidence interval for each cell line.
MSD assay
For total MCL1, ECL signal was recorded and converted to pg/mL as determined using calibration controls developed with MSD and using 4-parameter curve fit functions in MSD WorkBench software via 8-point standard dose range (0 pg/mL-10,000 pg/mL). The same procedure was used to convert the MCL1-BAK and MCL1-BIM dimer results to pg/mL, but with standard concentrations ranging from 0pg/mL to 50000 pg/mL. GAPDH results were recorded and reported as ECL and were used to normalize MCL1 and MCL1 dimers within each sample set. For graphical comparisons between analytes for a given cell line, each pg/mL dataset was normalized to be at 100% vehicle control and, e.g., 0% protein free.
Paclitaxel has been reported to down-regulate MCL1 protein levels { Wertz 2011}, in part, by an elevation of MCL 1E 3-ligase FBXW7 (which targets MCL1 for proteasome degradation). To confirm this observation, HCC70, MDA-MB-468 and HCC1806 TNBC cell line were treated with clinically relevant concentrations of paclitaxel (1 μm) for 4 hours { Gianni 1995}. Following paclitaxel treatment, the cells were incubated overnight and protein levels were determined. Comparison of paclitaxel treatment with vehicle control revealed elevated FBXW7 protein levels and reduced MCL1 protein levels (table 13 and fig. 1). Paclitaxel treatment also resulted in reduced protein levels of MCL1-BAK and MCL1-BIM dimers (table 13 and fig. 1). These results were observed across all three TNBC cell lines (n=3 bio-replicates).
HCC70, MDA-MB-468 and HCC1806 TNBC cell lines were treated with clinically relevant concentrations of paclitaxel (1 μm) for 4 hours { Gianni 1995}. In all three cell lines, paclitaxel treatment increased FBXW7 protein levels, decreased MCL1 protein levels, and decreased protein levels of MCL1-BAK and MCL1-BIM dimers. When HCC70, MDA-MB-468 and HCC1806 were titrated with paclitaxel doses (including protein-tuned C) max =1 μΜ) was pre-treated for 4 hours to simulate clinical exposure and then dose titrated for 72 hours to compound a, a Bliss synergy was observed [ ]>100)。
TABLE 13 percent change relative to vehicle after paclitaxel treatment (%)
a average normalized protein level (n=3 biological parallel assays)
To determine whether reduced MCL1 protein levels after paclitaxel treatment resulted in enhanced sensitivity to compound a, a Bliss synergy was used. HCC70, MDA-MB-468 and HCC1806 were titrated with paclitaxel doses (including protein-tuned C) max =1 μm) was pre-treated for 4 hours to simulate clinical exposure, and then exposed to dose titration of compound a. Cells were incubated for 72 hours and viability was determined using CTG reagents. Bliss synergy was observed across all three TNBC cell lines exposed in vitro to the combination of compound A and paclitaxel (Table 15). Bliss synergy scores greater than 100 are considered strong synergy { Prichard 1990}.
TABLE 14 Compound A+Taxol Bliss synergy score at 95% confidence interval
Table 15. Percent hcc70 inhibition results (n=5)
Paclitaxel (mu M)
Table 16 HCC70 synergistic results (95% CI)
Paclitaxel (mu M)
TABLE 17 percent MDA-MB-468 inhibition results(n=5)
Paclitaxel (mu M)
Table 18.MDA-MB-468 synergistic results (95% CI)
Paclitaxel (mu M)
Table 19.Hcc1806 percent inhibition results (n=5)
Paclitaxel (mu M)
TABLE 20 HCC1806 synergistic results (95% CI)
Paclitaxel (mu M)
Example 3: compound a was tested and targeted in a panel of breast cancer cell lines using a 72 hour in vitro proliferation assay Combination potential of agents and chemotherapy. The results of the combined tests are shown in Table 21
TABLE 21 in vitro combination test results
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
Example 4: compound a and BTK inhibition were tested in a panel of hematologic cancer cell lines using a 72 hour in vitro proliferation assay Combination potential of agents. The results of the combined test are shown in Table 22
TABLE 22 in vitro combination test results
/>
Example 5: compound a was tested in a panel of TNBC and NSCLC cancer cell lines using a 72 hour in vitro proliferation assay Combined potential with SN-38. The results of the combined tests are shown in Table 23
TABLE 23 in vitro combination test results
Cancer cell lines Bliss synergy score
HCC70(TNBC) 442
HCC1806(TNBC) 246
HCC1187(TNBC) 245
H522(NSCLC) 614
H820(NSCLC) 183
Example 6: in vivo testing of Compound A with paclitaxel in TNBC PDX model in anti-tumor efficacy study Combining potential (FIGS. 5 and 6)
Example 7: for evaluating compound a as monotherapy and in combination with anticancer therapy in the presence of a solid malignancy Safety, tolerability and pharmacokinetic phase 1a/b studies in subjects with tumors
A study will be performed to characterize the safety and tolerability of compound a as well as compound a in combination with an anti-cancer therapy in subjects with advanced solid malignancies.
Study design
This is a non-blind, multicentric, dose escalation and dose extending phase 1a/1b study for evaluating the safety, tolerability and PK profile of compound a, and for documenting any DLT (dose limiting toxicity), and for determining the MTD (maximum tolerated dose) and/or RP2D (recommended phase 2 dose) of compound a in subjects with advanced solid malignancies as monotherapy and in combination with anti-cancer therapies. RP2D will be at a dosage level with acceptable tolerability, exposure, efficacy and biomarker activity. The study will consist of phase 2, phase 1a (dose escalation), followed by phase 1b (dose extension):
stage 1a dose escalation: part A: dose escalation of Compound A as monotherapy
Stage 1b dose spread: part B: an optional disease-specific cohort of compound a in parallel with part a in combination with an anti-cancer therapy.
Part C: safety test of Compound A in combination with anticancer therapy after part A and part B (safety) runin) and expansion
Each part of the study will consist of screening, treatment and follow-up period. Screening will be performed 28 days prior to the first dose of study treatment, during which time eligibility and baseline characteristics of the subjects will be determined.
Part A: compound a phase 1 dose escalation as monotherapy
Subjects with advanced solid tumors who failed or were intolerant to standard therapy or for whom standard therapy was not present will be recruited successively to receive compound a as monotherapy at progressively higher dose levels.
Dose escalation will be performed using a dose escalation design based on the 3+3 rule.
Compound a will be administered orally for up to 105 weeks on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
Up to 6 cohorts (i.e., 6 dose levels), with 3 to 6 subjects each receiving increasing dose levels of compound a as monotherapy. The planned starting dose for compound a was 5mg, with target doses for the following 2 cohorts being 15mg and 50mg. Subsequent dose levels after the starting dose will be determined based on all available clinical data, including safety, tolerability and PK (pharmacokinetics) from the previous cohort and approved by SRT (safety review team), and possibly up to 300mg. In each subsequent dose increment, the dose level increase will be half-log (half-log) or less.
All subjects in the cohort will be followed for at least 21 days after the first dose of compound a, or after the subject has DLT during the first 21 days of study drug administration, the safety and tolerability of each dose level will be assessed by SRT.
The initial group for each dose consisted of 3 subjects. Dose escalation will be performed if no subject experiences DLT during the first 21 days of study drug administration. If 1 subject within the initial cohort of 3 subjects experienced DLT during the first 21 days of study drug administration, additional 3 subjects will be enrolled at the same dose level. If no DLT was observed in the additional 3 subjects, dose escalation will be performed. If 2 or more subjects experienced DLT during the first 21 days, the dose was decremented to a lower dose. MTD is the highest dose level where the incidence of DLT in the subjects was less than 33% during the first 21 days of study drug administration.
Treatment was performed for 21 days at any given queue using a consistent regimen to make the decision rule applicable.
Throughout the study, subjects with biopsied accessible malignant tumors may be subjected to an optional tumor biopsy. These subjects must agree and give individual, specific written agreements.
Dose escalation criteria
For any given cohort, the sponsor may choose to keep dosing, choose an intermediate dose, or stop study recruitment at any time based on a review of the primary safety and available PK and/or pharmacodynamic data.
Based on the review of relevant safety and available PK and/or pharmacodynamic data by SRT, the incrementing to higher dose queues will only be done in the absence of DLT and/or meeting any pre-specified stopping criteria. Dose escalation for subsequent cohorts with a magnitude greater than half-log would require confirmation from at least two-thirds of the majority of SRTs.
Dose limiting toxicity is defined as the following compound a-related events (after the first dose of compound a) that occurred within the first 21 days:
grade 4 hematotoxicity persists for more than 21 days
All compound a-associated grade 3 non-hematological toxicities lasted >7 days, and all compound a-associated grade 4 non-hematological toxicities were considered DLT regardless of duration
Part B: optional disease-specific cohort of compound a in parallel with part a in combination with anticancer therapy
During monotherapy dose escalation in part a, and prior to formal dose escalation of the sponsor-specified and approved disease-specific cohort characterized by combination therapy with compound a in part C, the sponsor may choose to specify and approve 1 or more of the following cohorts (which are aligned with those of part C for combination therapy with any previously evaluated dose of compound a in part a that is deemed safe and tolerable by SRT):
Queue B1: metastatic NSCLC (Compound A+docetaxel)
Queue B2: metastatic NSCLC (Compound A+ Sha Xituo bead monoclonal antibody goretinide)
Queue B3: metastatic TNBC (Compound A+docetaxel)
Queue B4: metastatic TNBC (Compound A+ Sha Xituo bead monoclonal antibody goretinide)
Queue B5: mSTS with nonspecific tissue structure (compound A+docetaxel and gemcitabine)
Each additional queue will consist of a single such population with a specific combination.
Part C: safety test and dose expansion of compound a in combination with other anticancer therapies
This is a non-blind 1B phase study with compound a administered in combination with other anti-cancer therapies in 1 or more of the 5 following disease-specific cohorts specified and approved by the sponsor after completion of parts a and B:
queue C1: metastatic NSCLC (Compound A+docetaxel)
Queue C2: metastatic NSCLC (Compound A+ Sha Xituo bead monoclonal antibody goretinide)
Queue C3: metastatic TNBC (Compound A+docetaxel)
Queue C4: metastatic TNBC (Compound A+ Sha Xituo bead monoclonal antibody goretinide)
Queue C5: mSTS with nonspecific tissue structure (compound A+docetaxel and gemcitabine)
RP2D will be at a dosage level with acceptable tolerance, exposure and biomarker activity.
Based on the population of clinical, safety, PK and pharmacodynamic data, SRT will recommend an initial dose of compound a for use in the combination for each cohort. A safety trial group of at least 3 subjects and no more than 6 subjects will be enrolled to ensure that the combination therapy is safe and tolerable in each subject population.
The safety trial will employ the same 3+3 design and dose escalation rules as part a and use the same DLT criteria and DLT evaluation window as part a to determine MTD and/or RP2D. A minimum of 6 subjects need to be treated at a dose level before the dose level can be extended. If the relevant disease-specific cohort from part B is explored under RP2D, those subjects may be counted and considered for safety trial group requirements.
The extension will include about 30 subjects, less any disease-specific related subjects treated with the same regimen explored in combination under part B and in any safety trial. For each disease-specific cohort (b1+c1, b2+c2, b3+c3, b4+c4, b5+c5), a minimum of 20 subjects under RP2D will be enrolled, including any from part B and/or safety trials.
Queue C1: combination with docetaxel in metastatic NSCLC following monotherapy for metastatic disease Compound A
Cohort C1 will evaluate safety and tolerability after single line therapy for metastatic disease, and define DLT and MTD and/or RP2D of compound a in combination with docetaxel in subjects with metastatic NSCLC.
Compound a will be administered on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
Docetaxel will be administered as 75mg/m in IV infusions over 1 hour on day 1 of each 21 day cycle 2 Is administered with the proviso that the subject's neutrophil count is allowable on the day of administration, in particular ≡1500 cells/mm 3
Treatment will last up to 105 weeks unless 1 or more disruption criteria are met.
Queue C2: taraxaco-Sha Xituo beads in metastatic NSCLC following monotherapy for metastatic disease Compound A of a combination of tikang
Cohort C2 will evaluate safety and tolerability following single line therapy for metastatic disease, and define DLT and MTD and/or RP2D of compound a in combination with Sha Xituo bead mab gautecan in subjects with metastatic NSCLC.
Compound a will be administered on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
Sha Xituo bead MAG-Geviracon will be administered as 10mg/kg in IV infusions once per week on days 1 and 8 of each 21 day cycle, provided that the subject's neutrophil count is allowed on the day of administration, in particular ≡1500 cells/mm on day 1 of any cycle 3 Or Absolute Neutrophil Count (ANC) at day 8 of any cycle ≡1000 cells/mm 3 . The first infusion should be administered over a period of 3 hours, wherein the subject is observed for signs or symptoms of infusion-related reactions during and for at least 30 minutes after infusion. If tolerised to a previous infusion, a subsequent infusion should be administered over 1 to 2 hours, with the subject being observed during and for at least 30 minutes after the infusion.
Treatment will last up to 105 weeks unless 1 or more disruption criteria are met.
Queue C3: combination of docetaxel in metastatic TNBC following single line therapy for metastatic disease Compound A
Cohort C3 will evaluate safety and tolerability after single line therapy for metastatic disease, and define DLT and MTD and/or RP2D of compound a in combination with docetaxel in subjects with metastatic TNBC.
Compound a will be administered on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
Docetaxel will be administered as 75mg/m in IV infusions over 1 hour on day 1 of each 21 day cycle 2 Provided that the subject's neutrophil count is allowed on the day of administration, in particular ≡1500 cells/mm 3
Treatment will last up to 105 weeks unless 1 or more disruption criteria are met.
Queue C4: against metastatic diseaseGolian with Sha Xituo bead mab in metastatic TNBC following single line therapy Compound A of kang combination
Cohort C4 will evaluate safety and tolerability following single line therapy for metastatic disease and define DLT and MTD and/or RP2D of compound a in combination with Sha Xituo bead mab govirukang in subjects with metastatic TNBC.
Compound a will be administered on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
Sha Xituo bead MAG-Geviracon will be administered as 10mg/kg in IV infusions once per week on days 1 and 8 of each 21 day cycle, provided that the subject's neutrophil count is allowed on the day of administration, in particular ≡1500 cells/mm on day 1 of any cycle 3 Or on day 8 of any cycle ANC.gtoreq.1000 cells/mm 3 . The first infusion should be administered over a period of 3 hours, wherein the subject is observed for signs or symptoms of infusion-related reactions during and for at least 30 minutes after infusion. If tolerised to a previous infusion, a subsequent infusion should be administered over 1 to 2 hours, with the subject being observed during and for at least 30 minutes after the infusion.
Treatment will last up to 105 weeks unless 1 or more disruption criteria are met.
Queue C5: metastatic soft tissue constructs with non-specific tissue structures that have not been previously treated for metastatic disease Malignant tumor of tissue
Cohort C5 will evaluate safety and tolerability, and define DLT and MTD and/or RP2D of compound a in combination with gemcitabine and docetaxel in subjects with soft tissue malignancies that were not previously treated.
Compound a will be administered on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
Gemcitabine will be administered as an IV infusion at a fixed dose rate of 900mg/m2 BSA over 90 minutes on days 1 and 8 of each 21 day cycle, with docetaxel being administered at 100mg/m2 BSA IV over 60 minutes on day 8.
Treatment will last up to 105 weeks unless 1 or more disruption criteria are met.
Duration of treatment
Study drug compound a will be administered for up to 105 weeks, or until disease progression, unacceptable toxicity, significant non-compliance with the study procedure or study drug, study discontinuation, withdrawal from the study, or other reasons (whichever occurs first).
Example 8: in vitro combinatorial screening for test Compound A with SN-38 in bladder and prostate cancer
The study used a 72 hour proliferation assay to evaluate the combined potential of compound a and SN-38 in a panel of bladder and prostate cancer cell lines. All tests were performed and reported by Horizon Discovery (cambridge, england). Compounds were tested for individual agent dose response in a 9x9 combination matrix with compound a. The combined results are ranked using the synergy scoring metrics.
Materials and methods
All cell lines and compounds (excluding compound a) were supplied and maintained by Horizon Discovery. Compound a was provided by gilid sciences company as Horizon Discovery.
TABLE 24 bladder and prostate cancer cell lines tested
TABLE 25 Compounds tested
Measurement
Cells were thawed from the liquid nitrogen storage state and expanded until dividing at their expected doubling time. Cells were seeded in growth medium in black 384-well tissue culture treatment plates and equilibrated via centrifugation. At the time of treatment, the "time zero" set of assay plates (which received no treatment) was collected and measured. The treated assay plates were incubated with the compounds (assayed in 3 replicates) for 3 days. After the required processing time, the Cell Titer Glow (plagmatogram) program and data point collection were performed via an automated process. Proprietary software from horizons was used to control and analyze the quality of the data.
Data analysis
Horizons utilized Growth Inhibition (GI) as a measure of cell growth. The GI percentage was calculated by applying the following test and formula:
if T < V_0:100 (1- (T-V_0)/V_0)
If T is greater than or equal to V_0:100 (1- (T-V_0)/(V-V_0))
Where T is the signal metric for the test article, V is the untreated/vehicle-treated control metric, and v—0 is the untreated/vehicle control metric at time zero (also colloquially referred to as T0 plate). The formula is derived from the growth inhibition calculations used in NCI-60 high throughput screening at the national cancer institute (National Cancer Institute).
A GI reading of 0% indicates no growth inhibition and will occur in the case where the T reading at 3 days is comparable to the V reading at the corresponding time period. 100% GI indicates complete growth inhibition (cell arrest), and in this case cells treated with the compound for 3 days will have the same end reading as T0 control cells. 200% GI indicates complete death (cytotoxicity) of all cells in the culture well, and in this case the T reading at 3 days will be lower than the T0 control (value near or at zero).
GI50 values from the growth inhibition measure are used to provide individual agent compound a activity. The maximum response observed was the highest growth inhibition measured with compound a. Both endpoints are reported as the mean of 20 independent results taken from a single agent compound a curve across the 20 combinations tested.
Horizons provide proprietary synergy scores based on the Loewe accumulation principle to characterize the strength of the synergistic interaction for the tested combinations. Horizons also generated synergistic results based on three standard models of combined effects: the highest individual agent (HSA), bliss, and Loewe accumulated as described above. All methods rely on a comparison of individual agent dose response curves to a combined matrix. For the purposes of this study report we focused on Horizon Synergy Score as it is a comprehensive measure of the combined effects. Any positive score reported indicates a synergistic interaction. The Bliss independent score, which is related to the internal method of assessing synergy, is included in the appendix as an alternative representation of the combined results.
Results
The combined test of compound a and SN-38 was determined in a 72 hour in vitro cell viability assay. Multiple synergy models were used to determine the combined activity of compound a and SN-38 (table 26). In vitro synergy of compound a and SN-38 was observed in multiple bladder cancer cell line models.
TABLE 26 Combined Activity of Compounds A+SN-38
Example 9: compound AF combined with Sha Xituo bead monoclonal antibody goretinide
In vivo Activity of anti-TNBC cancer cell line model MDA-MB-468.
Materials and methods
One hundred sixty-eight female athymic nude mice were inoculated in situ into a third fat pad (using matrigel) with 5.0X10-6 MDA-MB-468 cells. When the average tumor size reaches about 200mm 3, animals are randomized based on tumor volume. Dosing was started on study day 0.
TABLE 27 test Agents and treatment groups
Observation in life
General observations were performed daily. Body weight, tumor volume and clinical observations were recorded twice weekly.
Data analysis
Descriptive statistics are generated from the study data. The data is evaluated to determine if parametric or non-parametric analysis is appropriate. For parametric data, analysis of variance (ANOVA) was performed, followed by post hoc testing to determine significant differences between treatments, time points and/or groups. For non-parametric data, appropriate statistical analysis (e.g., kaplan-Meier survivinal, kruskal-Wallis One-way ANOVA, mann-Whitney or Wilcoxon Rank Sum, etc.) is performed.
Results
For MDA-MB-468 (TNBC) tumors, each mouse was dosed with 15mg/kg or 30mg/kg of Compound A (QD (2 day dosing/5 day dosing)) in combination with 200mg of Sha Xituo bead gorvelukant (IV QW (2 day dosing/1 day dosing)) for 6 weeks. Non-targeted ADC, h679-SN-38, was tested in combination with compound a as a control. Tumor volumes were determined for each treatment group (fig. 7).
Reference to the literature
Ashkenazi A,Fairbrother WJ,Leverson JD,Souers AJ.From basic apoptosis discoveries to advanced selective BCL-2family inhibitors.Nat Rev Drug Discov 2017;16(4):273-84。
Gianni L, kearns CM, giani A, capri G, vigano L, lacateli A, et al Nonlinear pharmacokinetics and metabolism of paclitaxel and its pharmacokinetic/pharmacodynamic relationships in humans J Clin Oncol1995;13 (1):180-90.
Juin P,Geneste O,Gautier F,Depil S,Campone M.Decoding and unlocking the BCL-2dependency of cancer cells.Nat Rev Cancer 2013;13(7):455-65。
Prichard MN,Shipman C,Jr.A three-dimensional model to analyze drug-drug interactions.Antiviral Res 1990;14(4-5):181-205。
Ruefli-Brasse A,Reed JC.Therapeutics targeting Bcl-2in hematological malignancies.Biochem J 2017;474(21):3643-57。
Wertz IE, kusam S, lam C, okamoto T, sandoval W, anderson DJ, et al Sensitivity to antitubulin chemotherapeutics is regulated by MCL and FBW7.Nature 2011;471 (7336):110-4.
Youle RJ,Strasser A.The BCL-2protein family:opposing activities that mediate cell death.Nat Rev Mol Cell Biol 2008;9(1):47-59。

Claims (37)

1. A method of treating cancer, comprising:
administering to a human patient in need thereof a therapeutically effective amount of an antibody-drug conjugate and a therapeutically effective amount of an MCL-1 inhibitor;
wherein the antibody-drug conjugate comprises an anti-Trop-2 antibody and an anti-cancer agent; and is also provided with
Wherein the MCL-1 inhibitor has the formula (I):
or a pharmaceutically acceptable salt thereof;
wherein R is 1 Is a 5-10 membered heteroaryl group containing 1-2 heteroatoms; wherein each heteroatom is independently selected from nitrogen, sulfur, and oxygen;
R 1 optionally independently selected from halogen, hydroxy, -CN, C 1-6 Alkyl, C 1-6 Haloalkyl, -OR a And C 3-6 1-3 substituents of cycloalkyl; and is also provided with
Each R 2 、R 3 、R 4 And R is 5 Independently hydrogen or C 1-6 An alkyl group;
R 6 hydrogen or halogen; and is also provided with
R a Independently hydrogen, C 1-6 Alkyl, C 2-6 Alkenyl and C 3-10 Cycloalkyl groups.
2. The method of claim 1, wherein the MCL-1 inhibitor is compound (a) or a pharmaceutically acceptable salt thereof:
3. the method of claim 1, wherein the MCL-1 inhibitor is selected from AMG-397, AMG-176, PRT-1419, and S64315.
4. A method according to any one of claims 1 to 3, wherein the anti-Trop-2 antibody is selected from hRS7, trop-2-XPAT and BAT-8003.
5. The method of any one of claims 1-4, wherein the anti-Trop-2 antibody is hRS7.
6. The method of any one of claims 1 to 5, wherein the anticancer agent is selected from the group consisting of Doxorubicin (DOX), epirubicin, morpholino doxorubicin (morpholino-DOX), cyano morpholino-doxorubicin (cyano morpholino-DOX), 2-pyrrolinyl-doxorubicin (2-PDOX), CPT, 10-hydroxycamptothecin, SN-38, topotecan, lurpetitacon, 9-aminocamptothecin, 9-nitrocamptothecin, taxane, geldanamycin, ansamycin, and epothilone.
7. The method of any one of claims 1 to 6, wherein the anti-cancer agent is SN-38.
8. The method of any one of claims 1 to 7, wherein the antibody-drug conjugate is salcetuximab goverikang.
9. The method of any one of claims 1 to 8, wherein the antibody-drug conjugate is salmeterol xinafoate gorboom and the MCL-1 inhibitor is compound a.
10. A method according to any one of claims 1 to 3, wherein the antibody-drug conjugate is dapobutan Shan Kangde lutecan.
11. The method of any one of claims 1 to 10, wherein the cancer is a Trop-2 expressing cancer.
12. The method of any one of claims 1 to 10, wherein the cancer is selected from breast cancer, cervical cancer, colorectal cancer, endometrial cancer, epithelial ovarian cancer, esophageal cancer, follicular thyroid cancer, gastric or gastroesophageal junction adenocarcinoma, head and neck cancer, lung cancer, hepatocellular carcinoma, non-small cell lung cancer, ovarian cancer, prostate cancer, renal cell cancer, small cell lung cancer, urothelial cancer, and urinary system cancer.
13. The method of any one of claims 1 to 12, wherein the cancer is selected from TNBC, hr+/HER2-BC, UC, NSCLC, SCLC, HNSCC and MIBC.
14. The method of any one of claims 1 to 13, wherein the cancer is metastatic.
15. The method of any one of claims 1 to 14, wherein the cancer is metastatic non-squamous non-small cell lung cancer (mNSCLC).
16. The method of any one of claims 1 to 14, wherein the cancer is metastatic triple negative breast cancer (mTNBC).
17. The method of any one of claims 1 to 14, wherein the cancer is a metastatic soft tissue malignancy with non-specific tissue architecture.
18. The method of any one of claims 1-17, wherein the human patient has received at least one additional therapy prior to treatment with the combination therapy of the MCL-1 inhibitor and the antibody-drug conjugate.
19. The method of any one of claims 1-18, wherein the human patient fails to respond to therapy with an anti-PD 1 or anti-PDL 1 agent prior to treatment with the combination therapy of the MCL-1 inhibitor and the antibody-drug conjugate.
20. The method of any one of claims 1 to 15, wherein a compound of formula (I) or a pharmaceutically acceptable salt thereof is administered simultaneously or separately with the conjugate.
21. The method of any one of claims 1 to 20, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered orally.
22. The method of any one of claims 1 to 21, wherein the amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered at a dose of about 5mg/kg, about 15mg/kg, or about 50 mg/kg.
23. The method of any one of claims 1 to 22, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered at a dose of about 5 mg/kg.
24. The method of any one of claims 1 to 23, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in a 21 day cycle, wherein administration is discontinued for 5 days after 2 days.
25. The method of any one of claims 1 to 24, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered for up to 105 weeks on days 1, 2, 8, 9, 15 and 16 of each 21 day cycle.
26. The method of any one of claims 1 to 25, wherein the antibody-drug conjugate is administered in the form of intravenous infusion.
27. The method of any one of claims 1 to 26, wherein the antibody-drug conjugate dose is administered on days 1 and 8 of each 21-day cycle.
28. The method of any one of claims 1-27, wherein the antibody-drug conjugate is administered at a dose of between about 4mg/kg and about 12 mg/kg.
29. The method of any one of claims 1-28, wherein the antibody-drug conjugate is administered at a dose selected from about 4mg/kg, about 6mg/kg, about 8mg/kg, about 10mg/kg, and about 12 mg/kg.
30. The method of any one of claims 1 to 29, further comprising radiation therapy.
31. The method of any one of claims 1 to 30, further comprising administering one or more additional therapeutic agents.
32. The method of claim 31, wherein the additional therapeutic agent is selected from the group consisting of a chemotherapeutic agent, a checkpoint inhibitor, and a BTK inhibitor.
33. The method of claim 32, wherein the checkpoint inhibitor is selected from the group consisting of an anti-PD-1 agent, an anti-PD-L1 agent, an anti-PD-1/PD-L1 interaction inhibitor, an anti-CTLA 4 agent, and an anti-TIGIT agent.
34. The method of any one of claims 32-33, wherein the checkpoint inhibitor is selected from the group consisting of nivolumab, pembrolizumab, atilizumab, pilizumab, and sapalimumab.
35. The method of claim 32 or 33, wherein the checkpoint inhibitor is selected from the group consisting of ipilimumab, lambrolizumab, tremelimumab, dulcis You Shan antibody, avermectin, tireli Li Youshan antibody, AB308, and domvanalimab.
36. The method of claim 32, wherein chemotherapy is performed with a chemotherapeutic agent; and wherein the chemotherapeutic agent is selected from docetaxel, p-paclitaxel, and gemcitabine.
37. The method of claim 32, wherein the BTK inhibitor is selected from the group consisting of acartinib, tiramitinib, zebutinib, and PCI-32765.
CN202280038962.9A 2021-06-11 2022-06-09 Combination of MCL-1 inhibitors with antibody drug conjugates Pending CN117396233A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/209,667 2021-06-11
US202263322509P 2022-03-22 2022-03-22
US63/322,509 2022-03-22
PCT/US2022/032816 WO2022261310A1 (en) 2021-06-11 2022-06-09 Combination mcl-1 inhibitors with anti-body drug conjugates

Publications (1)

Publication Number Publication Date
CN117396233A true CN117396233A (en) 2024-01-12

Family

ID=89467118

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280038962.9A Pending CN117396233A (en) 2021-06-11 2022-06-09 Combination of MCL-1 inhibitors with antibody drug conjugates

Country Status (1)

Country Link
CN (1) CN117396233A (en)

Similar Documents

Publication Publication Date Title
KR102454978B1 (en) 1-[4-bromo-5-[1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1 for the treatment of cancer associated with genetic abnormalities of platelet-derived growth factor receptor alpha Use of ,6-naphthyridin-3-yl]-2-fluorophenyl]-3-phenylurea and analogs
JP7014731B2 (en) Substituted aminopurine compounds, their compositions, and therapeutic methods using them.
TWI654979B (en) Method of treating cancer using TOR kinase inhibitor combination therapy
TW202038957A (en) Combination of antibody-drug conjugate and kinase inhibitor
TWI631949B (en) Methods for treating cancer using tor kinase inhibitor combination therapy
KR102271344B1 (en) Treatment of cancer with dihydropyrazino-pyrazines
CN114867622A (en) Combination therapy involving diaryl macrocycle compounds
Sclafani et al. Management of metastatic pancreatic cancer: Current treatment options and potential new therapeutic targets
CN104483407B (en) Micro-Imatinib assay method and the application in zero clinical trial phase in blood sample
CN117651554A (en) Combination of MCL-1 inhibitors and anticancer agents
US11931424B2 (en) Combination MCL-1 inhibitors with anti-body drug conjugates
CN117396233A (en) Combination of MCL-1 inhibitors with antibody drug conjugates
CN114533879A (en) Combination therapy for the treatment of cancer
US8709420B2 (en) Pharmaceutical combination of pazopanib and topotecan to treat neuroblastoma, osteosarcoma, and rhabdomyosarcoma in a human
CA3141072A1 (en) Methods and uses for treating cancer
EA045102B1 (en) APPLICATION OF 1-[4-BROMO-5-[1-ETHYL-7-(METHYLAMINO)-2-OXO-1,2-DIHYDRO-1,6-NAPHYRIDIN-3-YL]-2-FLUOROPHENYL]-3-PHENYLUREA AND ANALOGUES FOR THE TREATMENT OF CANCER ASSOCIATED WITH GENETIC DISORDERS IN THE PLATELET GROWTH FACTOR ALPHA RECEPTOR

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
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40106158

Country of ref document: HK