WO2024246287A1 - Medical use of a macrocyclic reversible btk inhibitor - Google Patents
Medical use of a macrocyclic reversible btk inhibitor Download PDFInfo
- Publication number
- WO2024246287A1 WO2024246287A1 PCT/EP2024/065039 EP2024065039W WO2024246287A1 WO 2024246287 A1 WO2024246287 A1 WO 2024246287A1 EP 2024065039 W EP2024065039 W EP 2024065039W WO 2024246287 A1 WO2024246287 A1 WO 2024246287A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- btk
- btk inhibitor
- subject
- reversible
- formula
- 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
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/4985—Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
- A61K31/52—Purines, e.g. adenine
- A61K31/522—Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57407—Specifically defined cancers
- G01N33/57426—Specifically defined cancers leukemia
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/154—Methylation markers
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/90—Enzymes; Proenzymes
- G01N2333/91—Transferases (2.)
- G01N2333/912—Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
- G01N2333/91205—Phosphotransferases in general
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/52—Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
Definitions
- the present invention relates to a medical use of a macrocyclic reversible BTK inhibitor and relates to a macrocyclic reversible BTK inhibitor for use in a method for treating a subject having a hyperproliferative disease.
- the subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor, preferably an irreversible BTK inhibitor, for treatment of the hyperproliferative disease.
- the method comprises a step of monitoring the subject over the course of therapy to determine whether the subject has a mutation in an endogenous gene encoding BTK.
- Kinases are enzymes that transfer a phosphate group from ATP to a protein. Kinases play an important role in regulating cellular functions such as cell proliferation, subcellular translocation, apoptosis, inflammation and metabolism (Attwood M.M. et al (2021) Nat Rev Drug Discov).
- the human kinome is composed of over 500 kinases. The development of smallmolecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy.
- BTK tyrosine kinase
- BCR B-cell receptor
- BTK small molecule inhibitors
- small molecule inhibitors such as the FDA approved covalent, irreversible, BTK inhibitors ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib
- CLL Chronic Lymphocytic Leukemia
- MCL Mantle Cell Lymphoma
- WM Macroglobulinemia
- SLL Small Lymphocytic Lymphoma SLL.
- BTK is also expressed and plays also pro-tumorigenic roles in several solid tumors (Xianhui Wang et al. 2021).
- BTK inhibition with ibrutinib or acalabrutinib inhibited cell growth (Kokabee et al 2015).
- BTK inhibitors have also showed inhibition of cellular proliferation and migration, and induced apoptosis and autophagy in glioblastoma cell lines (Wei et al., 2016; Wang et al., 2017).
- a drawback of the first generation BTK inhibitor, ibrutinib is that drug resistance in B- cell malignancies can develop when BTK acquires mutations at the cysteine at position C481 of the kinase domain. This mutation abrogates the covalent binding of ibrutinib hampering its efficacy.
- Quinquenel et al. performed a ‘snapshot’ screening to determine the prevalence of resistance mutations and found that the presence of the BTK mutation was significantly associated with subsequent CLL progression. The correlation between disease progression, and the emergence and temporal dynamics of the most common resistance inducing C481 S BTK mutation have been determined for CLL patients receiving single-agent Ibrutinib treatment (Bodor et al.
- Second-generation covalent, irreversible, BTK inhibitors which include acalabrutinib, zanubrutinib, and tirabrutinib, offer greater BTK selectivity and therefore limited off-target toxicity. These inhibitors, however, do not overcome resistance by C481 mutation.
- BTK inhibitors To treat patients with relapsed CLL having C481 BTK mutations non-covalent, reversible, BTK inhibitors have been developed including LOXO-305 (pirtobrutinib) and ARQ-531 (nemtabrutinib) and vecabrutinib. These agents do not require covalent binding to the BTK C481 residue and effectively inhibit both wild type and mutant BTK with C481 substitutions.
- non-covalent BTK inhibitors including pirtobrutinib (LOXO-305), ARQ-351 and vecabrutinib, inhibited B-cell- receptor signaling in BTK C481 -mutant cell and animal models.
- phase 1-2 clinical trial of pirtobrutinib showed promising efficacy for patients with B-cell cancer who had previously been treated with covalent BTK inhibitors (with 62% of patients with CLL having a response), including patients with or without BTK C481 mutations (with a response occurring in 71 % and 66% of the patients, respectively) (Wang et al. (2022) N. Engl. J. Med. 386, 735-43).
- Pirtobrutinib was first disclosed in WO2017/103611 , ARQ-531 was disclosed in WO2017/111787 and vecabrutinib is disclosed in WO2013/185084. Further reversible BTK inhibitors are disclosed in WO2017/046604, , WQ2020/015735, WQ2020/239124, WQ2021/093839, WQ2020/043638, WO2013/067274, WO2018097234, WO2013/010380, W02016/161570, WO2016/161571 , WO2016/106624, WO2016/106625, WO2016/106626, WO2016106623, WO2016/106628 and WO2016/109222.
- W is an aryl group having 6-10 carbon or a heteroaryl group having 1-5 carbon; wherein any said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;
- V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R 1v )-NH-C(O)-, -CH(R 1v )- ;
- R 1v is hydrogen or (1 -2C)alkyl
- U is an aryl group having 6-10 carbon or an heteroaryl group having 1 -5 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl, alkoxy, cycloalkyl and heterocycloalkyl group is optionally and independently substituted with one, two or three halogen; wherein R 2 is of Formula (I l-a) to (ll-f) selected from the group consisting of:
- a method for treating a subject having a hyperproliferative disease preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor
- the method comprising the steps: (a) monitoring the subject, preferably at predetermined intervals of time, over the course of the therapy for treatment of the hyperproliferative disease to determine whether the subject has a mutation in an endogenous gene encoding BTK that results in resistance or provides a risk of resistance of the subject to the therapy, preferably resistance of the subject to the irreversible BTK inhibitor, preferably the subject has a mutation in an endogenous gene encoding BTK that results in expression of a BTK protein with a modification at an amino acid position corresponding to amino acid position 481 of the amino acid sequence according to SEQ ID NO: I; and
- step (b) if the subject has shown said mutation, administering to the subject a reversible BTK inhibitor, in particular if the subject has the BTK modification corresponding to amino acid position 481 identified in step (a), wherein the reversible BTK inhibitor is a compound according to the invention having any one of Formula (l-a) to (l-h).
- a method for treating a subject having a hyperproliferative disease preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor
- the method comprises: optionally stopping the administration of the irreversible BTK inhibitor, and administering to said subject an effective amount of a reversible BTK inhibitor, wherein the reversible BTK inhibitor is a compound according to the invention having any one of Formula (I- a) to (l-h).
- a reversible BTK inhibitor for use in a method according to the invention which is a compound of any one of Formula (l-a) to (l-h).
- a reversible BTK inhibitor according to the invention in the manufacture of a medicament for treating a subject having a hyperproliferative disease, preferably treating a subject having a B-cell hematological malignancy.
- the present inventors have surprisingly found that the reversible BTK inhibitor according to the invention is very effective in inhibiting BTK mutants, e.g. in inhibiting B-cell lymphoma cells comprising two separate cell pools each containing a different mutation in BTK. Additionally, the inventors have found that the reversible BTK inhibitor according to the invention is very effective in treating subjects having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor.
- a hyperproliferative disease preferably a B-cell hematological malignancy
- a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor.
- composition as used herein has its conventional meaning and refers to a composition which is pharmaceutically acceptable.
- pharmaceutically acceptable has its conventional meaning and refers to compounds, material, compositions and/or dosage forms, which are, within the scope of sound medical judgment suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
- effective amount refers to an amount of the compound of the invention, and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventive effect when administered to a subject suffering from a BTK-mediated disease or disorder.
- effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount.
- a “subject” is a human or non-human mammal. In one embodiment, a subject is a human.
- the term “controlling” is intended to refer to all processes wherein there may be a slowing, interrupting, arresting or stopping of the progression of the diseases and conditions affecting the mammal. However, “controlling” does not necessarily indicate a total elimination of all disease and condition symptoms, and is intended to include prophylactic treatment.
- excipient as used herein has its conventional meaning and refers to a pharmaceutically acceptable ingredient, which is commonly used in the pharmaceutical technology for preparing a granulate, solid or liquid oral dosage formulation.
- salt as used herein has its conventional meaning and includes the acid addition and base salts of the compound of the invention.
- solvate as used herein has its conventional meaning.
- One or more compounds of the invention or the pharmaceutically acceptable salts thereof may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
- “Solvate” means a physical association of a compound of this invention with one or more solvent molecules. This physical association Involves varying degrees of ionic and covalent bonding. Including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
- “Solvate” encompasses both solution-phase and isolatable solvates. Examples of suitable solvates include ethanolates, methanolates, and the like.
- “Hydrate” is a solvate wherein the solvent molecule is H2O and includes any hydrate of the compound or the salt of said compound.
- treatment has its conventional meaning and refers to curative, disease controlling, palliative and prophylactic treatment.
- unit dosage form has its conventional meaning and refers to a dosage form which has the capacity of being administered to a subject, preferably a human, to be effective, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose comprising the therapeutic agent, i.e. the compound of the invention.
- BTK Bruton's Tyrosine Kinase
- BTK Bruton's tyrosine kinase
- Src-related Tec family of protein kinases which are a large subset of kinases which play a central role in the regulation of a wide variety of cellular signaling processes.
- BTK plays a key role in the B-cell receptor signaling and a critical role in the regulation of survival, proliferation, activation and differentiation of B- lineage cells.
- BTK small molecule inhibitors
- small molecule inhibitors such as the FDA approved irreversible BTK inhibitors ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib
- CLL Chronic Lymphocytic Leukemia
- MCL Mantle Cell Lymphoma
- WM Macroglobulinemia
- SLL Small Lymphocytic Lymphoma
- BTK inhibitor as used herein has its conventional meaning and refers to an inhibitor for BTK.
- a BTK inhibitor may be a small molecule inhibitor.
- Inhibitors may be irreversible inhibitors, such as by forming a covalent bond, and may be reversible inhibitors, which may form a reversible interaction with BTK.
- covalent BTK inhibitor has its conventional meaning and refers to a BTK inhibitor that reacts with its target protein (BTK) to form a covalent complex in which the protein has lost its function.
- BTK target protein
- Covalent inhibitors can be reversible or irreversible, depending on the rate of the reverse reaction.
- 'covalent inhibitor' and 'irreversible inhibitor' are often used interchangeably.
- irreversible BTK inhibitor has its conventional meaning and refers to a BTK inhibitor that possesses an off-rate that is slow relative to the rate of re-synthesis of the target protein (BTK) in vivo, so that once the target protein is inhibited, it does not regain activity.
- rate of re-synthesis has its conventional meaning and refers to the rate at which a cell and/or organism replaces a protein target with freshly synthesized functional protein.
- the re-synthesis rate defines the rate at which an irreversibly inhibited protein target will recover activity in vivo, once the inhibitor is no longer present.
- reversible BTK inhibitor has its conventional meaning and refers to a BTK inhibitor that inactivates the BTK enzyme through non-covalent, transcient, interactions. Unlike an irreversible inhibitor, a reversible inhibitor can dissociate from the enzyme.
- mutant-BTK has its conventional meaning and refers to mutations of BTK. Mutations of BTK may be referred to by an altered or modified amino acid target (such as C as single-letter amino acid code for cysteine) at a certain position of the BTK structure (such as 481). Additionally, the amino acid substitution at the modification position may be referred to by an additional amino acid single-letter amino acid code, such as C481 S for serine substitution and C481T for threonine substitution of cysteine at the 481 position.
- an altered or modified amino acid target such as C as single-letter amino acid code for cysteine
- an additional amino acid single-letter amino acid code such as C481 S for serine substitution and C481T for threonine substitution of cysteine at the 481 position.
- modification has its conventional meaning and refers to modification of a sequence of amino acids of a polypeptide, protein or a sequence of nucleotides in a nucleic acid molecule and includes deletions, insertions, and replacements of amino acids and nucleotides, respectively.
- the term "resistance to BTK inhibitor” as used herein has its conventional meaning and refers to a BTK inhibitor, which shows a reduction in effectiveness, after being effective initially in treating a hyperproliferative disease, in particular a B-cell hematological malignancy.
- a drawback of the currently approved irreversible inhibitors is that patients treated with these inhibitors can develop drug resistance when BTK proteins with variations at the catalytic site are not able to bind efficiently to irreversible inhibitors in patients. This is a rather common event in patients treated with irreversible inhibitors and who experience relapse.
- a major mechanism for the acquired resistance is the emergence of BTK cysteine 481 (C481) mutations. These mutations hamper binding of irreversible inhibitors such as ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib which form a covalent bond with this amino acid.
- valine 416 V416)
- A428 alanine 428
- M437 methionine 437
- T474 threonine 474
- L528 leucine 528
- drug resistance is meant the major cause of cancer treatment failure. While a treatment may be effective initially, the heterogeneity of cancer and its ability to adapt can allow the cancer to become resistant to the treatment and regrow.
- relapse as used herein has the conventional meaning and refers to evidence of disease progression in a patient who has previously achieved criteria of a complete response or partial remission.
- disease progression has the conventional meaning and refers to a measured increase in tumor size or tumor burden.
- recurrent or refractory form of a hyperproliferative disease has the conventional meaning and refers to a particular cancer that is resistant, or non-responsive to therapy with a particular therapeutic agent.
- a cancer can be refractory to therapy with a particular therapeutic agent either from the onset of treatment (i.e., non-responsive to initial exposure to the therapeutic agent), or as a result of developing resistance to the therapeutic agent, either over the course of a first treatment period with the therapeutic agent or during a subsequent treatment period with the therapeutic agent.
- the term ‘’acquired drug resistance” as used herein has the conventional meaning and refers to resistance of a drug caused by mutations in the target protein. The mutations in the target protein hamper the binding of drug resulting in a regrowth of the tumor
- clinical drug resistance refers to growth of a tumour while the patient is on treatment, that develops following after an initial clinical benefit (a clinical response or prolonged stable disease).
- wt-BTK or “WT-BTK” or “BTK WT ” as used herein has its conventional meaning and refers to wild-type Bruton’s Tyrosine Kinase.
- a wild-type BTK has the regular meaning of a phenotype of the typical form of BTK as it occurs in nature. Originally, the wild-type was conceptualized as a product of the standard "normal” allele at a locus, in contrast to that produced by a non-standard, "mutant” allele.
- microcycle as used herein has its conventional meaning and refers to a part of a molecule containing a ring consisting of 12 or more ring atoms forming said ring. In an example, a twelve membered ring consist of 12 atoms forming said ring.
- binding affinity as used herein has its conventional meaning and refers to the equilibrium dissociation constant which is an inverse measure of the affinity of a proteinligand (small molecule) pair under equilibrium conditions.
- the value of KD is mathematically equivalent to the ratio kotr/kon (or kd/k a ) measured using Surface Plasmon Resonance (SPR).
- association rate constant or “on-rate (k on or k a )” as used herein has its conventional meaning and refers to a second-order rate constant that quantifies the rate at which a free ligand and free protein combine (through collisional encounters) to form a binary proteinligand complex.
- dissociation rate constant or “off-rate (k O ft or kd)” as used herein has its conventional meaning and refers to a first-order rate constant that quantifies the rate at which a binary protein-ligand complex dissociates to the free ligand and free protein.
- target residence time tau (r) has its conventional meaning and refers to the time a compound resides on its target.
- Target residence time (T) can be determined according to the method as described below in the experimental section.
- IC50 as used herein has its conventional meaning and refers to the concentration of a substance that results in a 50% effect on some measure of biochemical and/or cellular function or substance-target binding interaction.
- pICso as used herein has its conventional meaning and refers to the negative logarithm of the IC50 in molar concentration.
- G/50 has its conventional meaning and refers to the concentration of a substance that inhibits cell growth by 50%.
- LD50 refers to the concentration of a substance that results in 50% cell death.
- a bicyclic ringsystem refers to heterocyclic (heterocyclyl) groups, to cyclic groups having carbon groups only, i.e. without hetero atoms, within the cycle, and to combinations of a heterocyclic (heterocyclyl) group and a cyclic group having carbon groups only, i.e. without hetero atoms, within the cycle.
- a monocylic ringsystem refers both to a heterocyclic (heterocyclyl) group, and to a cyclic group having carbon groups only, i.e. without hetero atoms, within the cycle.
- heterocyclic (heterocyclyl) group refers to both heteroaryl groups and heterocycloalkyl groups.
- a heterobicyclic group refers to a bicyclic group having one or more heteroatoms, which is saturated, partially unsaturated or unsaturated.
- aromatic groups include aromatic carbocyclic ring systems (e.g. phenyl) and fused polycyclic aromatic ring systems (e.g. naphthyl and 1 , 2,3,4- tetrahydronaphthyl).
- alkyl refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond having the specified number of carbon atoms.
- an alkyl group contains, for example, from 1 to 6 carbon atoms (1 - 6C)Alkyl or from 1 to 3 carbon atoms (1 -3C)Alkyl.
- alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl.
- an alkyl group is linear. In another embodiment, an alkyl group is branched.
- alkyl includes both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbon atoms; for example, “(1-6C)Alkyl” includes all of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
- Alkylene refers to both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbons, and having two terminal end chain attachments; for example, the term “A-C4 alkylene-B” represents, for example, A-CH2-CH2-CH2-CH2-B, A-CH2- CH 2 -CH(CH 3 )-CH 2 -B, A-CH 2 -CH(CH 2 CH 3 )-B, A-CH 2 -C(CH 3 )(CH 3 )-B, and the like.
- alkylcarbonyl refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond attached to a carbonyl group, wherein the aliphatic hydrocarbon group has the specified number of carbon atoms.
- an alkyl group or aliphatic hydrocarbon group contains, for example, from 1 to 6 carbon atoms (1 -6C)Alkyl or from 1 to 3 carbon atoms (1 -3C)Alkyl.
- alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl.
- an alkyl group is linear. In another embodiment, an alkyl group is branched.
- Cycloalkyl means a cycloalkyl group having the recited number of carbon atoms, with the same meaning as previously defined, such as cyclopropyl, cyclobutyl, or cyclopentyl.
- Cycloalkyl refers to a cycloalkyl-group represented by an indicated number of carbon atoms; for example “(3-6C)cycloalkyl” includes cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
- Heterocycloalkyl means a cycloalkyl group having the recited number of carbon atoms, and 1-3 heteroatoms selected from N, O and/or S, with the same meaning as previously defined.
- Haloalkyl means a branched or unbranched alkyl group having the recited number of carbon atoms, in which one and up to all hydrogen atoms are replaced by a halogen; halogen is as defined herein.
- branched or straight chained haloalkyl groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl and n- butyl substituted independently with one or more halogens, e.g., fluoro, chloro, bromo and iodo.
- a halo(1 -3C)alkyl means a branched or unbranched alkyl group having 1 ,2, or 3 carbon atoms, in which at least one hydrogen atom is replaced by a halogen.
- haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 1 - fluoroethyl, 2-fluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, and perfluoro-n-propyl.
- Alkoxy means an alkoxy group having the recited number of carbon atoms, the alkyl moiety having the same meaning as previously defined, e.g., "Alkoxy” refers to an alkyl-O-group represented by a linear or branched alkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "(1 -6C)Alkoxy” includes -OCH3, -O-CH2CH3, - OCH(CH 3 ) 2 , -O(CH 2 ) 5 CH3, and the like.
- Cycloalkoxy means a cycloalkyl group having the recited number of carbon atoms, with the same meaning as previously defined, attached via a ring carbon atom to an exocyclic oxygen atom, such as cyclopropoxyl, cyclobutoxyl,or cyclopentoxyl.
- Cycloalkoxy refers to a cycloalkyl- O-group represented by a cycloalkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "(3-6C)cycloalkoxy” includes cyclopropyl-O-, cyclobutyl- O-, cyclopentyl-O-, or cyclohexyl-O-.
- Heterocycloalkoxy means a cycloalkyl group having the recited number of carbon atoms, and 1-3 heteroatoms selected from N, O and/or S, with the same meaning as previously defined, attached via a ring carbon atom to an exocyclic oxygen atom.
- alkyl groups are unsubstituted or substituted with 1 to 3 substituents on each carbon atom.
- compositions comprising components A and B
- the only enumerated components of the composition are A and B, and further the claim should be interpreted as including equivalents of those components.
- indefinite article “a” or “an” does not exclude the possibility that more than one of the element or component are present, unless the context clearly requires that there is one and only one of the elements or components.
- the indefinite article “a” or “an” thus usually means “at least one”.
- Figure 1 Map of clinically documented BTK mutants.
- Figure 2 Proliferation assay of single compounds (monotherapy) in wt-BTK TMD8 cells.
- Figure 3 Proliferation assay of single compounds (monotherapy) in BTK C481 S TMD8 cells.
- Figure 4 Proliferation assay of single compounds (monotherapy) in BTK T474I TMD8 cells.
- Figure 5 Proliferation assay of single compounds (monotherapy) in BTK T474I/C481 S TMD8 cells.
- Figure 6 Proliferation assay of single compounds (monotherapy) in BTK V416L TMD8 cells.
- Figure 7 Proliferation assay of single compounds (monotherapy) in BTK L528W TMD8 cells.
- Figure 8 Proliferation assay of single compounds (monotherapy) in a mixture of BTK
- FIG. 9 Proliferation assay of single compounds (monotherapy) in a mixture of BTK C481 S TMD8 cells and BTK T474I TMD8 cells.
- Figure 10A Washout pirtobrutinib and compound A in wt-BTK GripTite 293 MSR cells.
- Figure 10B Quantification of washout pirtobrutinib and compound A in wt-BTK GripTite 293 MSR cells.
- Figure 11 A Washout pirtobrutinib and compound A in BTK C481 S GripTite 293 MSR cells.
- Figure 11 B Quantification of washout pirtobrutinib and compound A in BTK C481 S GripTite 293 MSR cells.
- Figure 12A Washout pirtobrutinib and compound A in BTK T474I GripTite 293 MSR cells.
- Figure 12B Quantification of washout pirtobrutinib and compound A in BTK T474I GripTite 293 MSR cells.
- Figure 13A Washout pirtobrutinib and compound A in BTK T474I/C481 S GripTite 293 MSR cells.
- Figure 13B Quantification of washout pirtobrutinib and compound A in BTK T474I/C481 S GripTite 293 MSR cells.
- Figure 14A Washout pirtobrutinib and compound A in BTK M437R GripTite 293 MSR cells.
- Figure 14B Quantification of washout pirtobrutinib and compound A in BTK M437R GripTite 293 MSR cells.
- Figure 15A Western blot Dose Response Curve results of compound A in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
- Figure 15B Western blot Dose Response Curve results of pirtobrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
- Figure 15C Western blot Dose Response Curve results of ibrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
- Figure 15D Western blot Dose Response Curve results of acalabrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
- Figure 15E Western blot Dose Response Curve results of nemtabrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
- Figure 15F Western blot Dose Response Curve results of ibrutinib, acalabrutinib, pirtobrutinib, nemtabrutinib and compound A in BTK M437R expressing 293 cells.
- FIG 16 Ibrutinib resistance mutations can be detected before clinical relapse.
- samples before relapse were analyzed retrospectively to determine the interval of time between mutation detection and clinical relapse.
- An initial clone could be detected at an estimated median of 9.3 months before relapse.
- UPN unique patient number (Woyach et al. 2017).
- the present inventors have surprisingly established that the macrocyclic reversible BTK inhibitor according to the invention is very effective in inhibiting BTK mutants, e.g. in inhibiting B- cell lymphoma cells comprising two separate cell pools each containing a different mutation in BTK. Additionally, the inventors have found that the reversible BTK inhibitor according to the invention is very effective in treating subjects having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor.
- a hyperproliferative disease preferably a B-cell hematological malignancy
- BTK Bruton’s tyrosine kinase
- compounds according to the invention provide an improved reversible binding activity towards wild-type BTK and/or BTK mutants.
- the compounds according to the invention have any one of Formula (l-a) to (l-h), which contains a macrocyclic moiety, in combination with BTK specific pharmacophores (e.g. based on ligands for binding to BTK) to provide a binding activity towards wild-type BTK and/or BTK mutants through improved reversible binding.
- BTK specific pharmacophores e.g. based on ligands for binding to BTK
- the reversible BTK inhibition compounds according to the invention have any one of Formula (l-a) to (l-h), which are extensively described in co-pending PCT application PCT/EP2021/085641 and in PCT application PCT/EP2022/085765.
- the reversible BTK inhibitors according to the present invention include the (macrocyclic) BTK inhibition compounds of the co-pending PCT application PCT/EP2021/085641 and of the PCT application PCT/EP2022/085765 and are incorporated by reference, including the exemplary compounds having sub-formula 1 - 226 and the described syntheses routes for manufacturing the compounds having any one of Formula (l-a) to (l-h).
- the reversible BTK inhibitor is a compound according to Formula (l-a) or (l-b) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound comprises a bicyclic scaffold selected from: -b)
- R 1 is any one of: wherein R 2w is selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any said alkyl or alkoxy group is optionally and independently substituted with one, two or three fluoro;
- R 3u is selected from hydrogen, halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3- 6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R 2 is selected from the group consisting of:
- any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with hydroxy, methyl, acetyl or methoxy; wherein R 3 and R 4 together represent a linker having Formula selected from the group consisting of: whereby the T ⁇ * marks the position of R 3 in any one of Formula l-a to l-b, and whereby the marks the position of R 4 in any one of Formula I l-a to I l-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CD3, (1 -4C)alkyl, (1-5C)alkoxy, (3- 6C)cycloalkyl, (3-6C)cycloalkoxy and (1 -6C)alkylcarbonyl; wherein any of said al
- the irreversible BTK inhibitor which the subject has received, is selected from the group consisting of: ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, spebrutinib, branebrutinib, evobrutinib, remibrutinib, tolebrutinib, orelabrutinib, elsubrutinib, edralbrutinib, ACP-5862, and pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
- the irreversible BTK inhibitor is ibrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
- the irreversible BTK inhibitor is acalabrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
- the irreversible BTK inhibitor is zanubrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
- the irreversible BTK inhibitor is tirabrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
- SEQ ID NO: 1 tyrosine-protein kinase BTK [Homo sapiens] 10 20 30 40 50 60
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the protein modification is C481 S.
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 of the amino acid sequence according to SEQ ID NO: I, wherein the modification is a substitution of cysteine to an amino acid selected from leucine, isoleucine, valine, alanine, glycine, methionine, serine, threonine, phenylalanine, tryptophan, lysine, arginine, histidine, proline, tyrosine, asparagine, glutamine, aspartic acid and glutamic acid at amino acid position 481 of the BTK protein.
- the modification is a substitution of cysteine to an amino acid selected from leucine, isoleucine, valine, alanine, glycine, methionine, serine, threonine, phenylalanine, tryptophan, lysine, arginine, histidine, proline, tyros
- the modification is a substitution of cysteine to serine at amino acid position 481 of the BTK protein.
- the subject is identified as having one or more mutations in a gene that results in expression of a BTK protein with a modification at one or more amino acid positions selected from the group consisting of: 416, 428, 437, 474, 481 and 528.
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 416 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is V416L.
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 428 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is A428D.
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 437 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is M437R.
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 474 of the amino acid sequence set according to SEQ ID NO: I, more preferably wherein the mutant modification is T474L
- the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 528 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is L528W.
- prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, ibrutinib before starting a new treatment option for managing the same condition.
- a hyperproliferative disease preferably a B-cell hematological malignancy
- BTK tyrosine kinase
- prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, acalabrutinib before starting a new treatment option for managing the same condition.
- a hyperproliferative disease preferably a B-cell hematological malignancy
- BTK tyrosine kinase
- prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, zanubrutinib before starting a new treatment option for managing the same condition.
- a hyperproliferative disease preferably a B-cell hematological malignancy
- BTK tyrosine kinase
- prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, tirabrutinib before starting a new treatment option for managing the same condition.
- a hyperproliferative disease preferably a B-cell hematological malignancy
- BTK tyrosine kinase
- the irreversible BTK inhibitor is administered during the prior therapy in an amount in the range of 70 - 750 mg / day and / or wherein the irreversible BTK inhibitor is administered using one or more unit doses having an amount in the range of 70 - 750 mg / unit dose.
- the irreversible BTK inhibitor is administered during the prior therapy daily at a dose selected from the group consisting of 70 mg, 100 mg, 140 mg, 160 mg, 200 mg, 280 mg, 320 mg, 420 mg, 480 mg, 560 mg per day.
- the irreversible BTK inhibitor is administered during the prior therapy twice daily at a dose of 80 mg, 100 mg or 160 mg..
- maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, ibrutinib.
- a hyperproliferative disease preferably a B-cell haematological malignancy
- BTK tyrosine kinase
- maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, acalabrutinib.
- BTK Bruton’s tyrosine kinase
- maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, zanubrutinib.
- maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, tirabrutinib.
- BTK tyrosine kinase
- the methods for maintenance therapy comprise treating a B-cell hematological malignancy with a covalent and/or irreversible BTK inhibitor for an initial treatment period, followed by a maintenance therapy regimen.
- the methods for maintenance therapy comprise treating a B-cell haematological malignancy with a covalent and/or irreversible BTK inhibitor for a period of six months or longer, such as, for example, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, 13 months, 14 months, 15 months, 16 months, 17 months, 18 months, 19 months, 20 months, 21 months, 22 months, 23 months, 24 months, 25 months, 26 months, 27 months, 28 months, 29 months, 30 months, 31 months, 32 months, 33 months, 34 months, 35 months, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years or longer.
- a covalent and/or irreversible BTK inhibitor for a period of six months or longer, such as, for example, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, 13 months, 14 months, 15 months, 16 months, 17 months, 18 months, 19 months, 20 months, 21 months, 22 months, 23 months, 24 months
- the subject is monitored over the course of the therapy with a BTK inhibitor for treatment of the hyperproliferative disease to determine whether the subject has a mutation in an endogenous gene encoding BTK that results in or provides a risk of resistance of the subject to the therapy, preferably resistance of the subject to the irreversible BTK inhibitor.
- the subject is monitored at predetermined intervals of time over the course of the therapy for treatment of the hyperproliferative disease, wherein the predetermined interval of time is every week, every 2 weeks, every 3 weeks, every month, every 2 months, every 3 months, every 4 months, every 5 months, every 6 months, every 7 months, every 8 months, every 9 months, every 10 months, every 11 months, every year following the first administration of the covalent and/or irreversible BTK inhibitor.
- the treatment which is monitored is in particular a treatment of administration of an irreversible BTK inhibitor.
- testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 481 of the BTK protein.
- PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 481 of the BTK protein.
- the method comprises seguencing the amplified nucleic acids.
- testing comprises performing deep seguencing of nucleic acids that encode amino acid position 481 of the BTK protein.
- testing comprises contacting the nucleic acids with a seguence specific nucleic acid probe, wherein the seguence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 481 ; and (b) does not bind to nucleic acids encoding the wild-type BTK having cysteine at amino acid position 481 .
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 416 of the BTK protein.
- PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 416 of the BTK protein.
- the method comprises sequencing the amplified nucleic acids.
- testing comprises performing deep sequencing of nucleic acids that encode amino acid position 416 of the BTK protein.
- testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 416; and (b) does not bind to nucleic acids encoding the wild-type BTK having valine at amino acid position 416.
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 428 of the BTK protein.
- PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 428 of the BTK protein.
- the method comprises sequencing the amplified nucleic acids.
- testing comprises performing deep sequencing of nucleic acids that encode amino acid position 428 of the BTK protein.
- testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 428; and (b) does not bind to nucleic acids encoding the wild-type BTK having alanine at amino acid position 428.
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 437 of the BTK protein.
- PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 437 of the BTK protein.
- the method comprises sequencing the amplified nucleic acids.
- testing comprises performing deep sequencing of nucleic acids that encode amino acid position 437 of the BTK protein.
- testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 437; and (b) does not bind to nucleic acids encoding the wild-type BTK having methionine at amino acid position 437.
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 474 of the BTK protein.
- PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 474 of the BTK protein.
- the method comprises sequencing the amplified nucleic acids.
- testing comprises performing deep sequencing of nucleic acids that encode amino acid position 474 of the BTK protein.
- testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 474; and (b) does not bind to nucleic acids encoding the wild-type BTK having threonine at amino acid position 474.
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 528 of the BTK protein.
- PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 528 of the BTK protein.
- the method comprises sequencing the amplified nucleic acids.
- testing comprises performing deep sequencing of nucleic acids that encode amino acid position 528 of the BTK protein.
- testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 528; and (b) does not bind to nucleic acids encoding the wild-type BTK having tryptophane at amino acid position 528.
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- the sample for use in the methods contains one or more tumor cells from the subject. In some embodiments, the sample for use in the methods contains cell free DNA (cfDNA). In some embodiments, the sample for use in the methods contains circulating tumor DNA (ctDNA). In some embodiments, the sample for use in the methods contains tumor derived extracellular vesicles (EV). In some embodiments, the sample is a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate.
- cfDNA cell free DNA
- ctDNA circulating tumor DNA
- EV tumor derived extracellular vesicles
- the sample is a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate.
- the nucleic acids used in the method is isolated from a tumor cell sample from the subject. In some embodiments of the methods, the nucleic acids used in the method are isolated from extracellular vesicles (EV) isolated from the subject. In some embodiments of the method the nucleic acids used in the method are isolated from a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate from the subject.
- EV extracellular vesicles
- the nucleic acids for use in the assay are RNA or DNA. In some embodiments of the methods, the nucleic acids for use in the assay are genomic DNA. In some embodiments of the methods, the nucleic acids for use in the assay are total RNA. In some embodiments of the methods, the nucleic acids for use in the assay are messenger RNA (mRNA).
- mRNA messenger RNA
- the nucleic acids for use in the assay are complementary DNA (cDNA).
- the method further comprises isolating mRNA from the RNA sample. In some embodiments of the methods, the method further comprises reverse transcription of the total RNA, or mRNA, into cDNA.
- testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 481 ; and (b) does not bind to nucleic acids encoding the wild-type BTK having cysteine at amino acid position 481.
- testing comprises PCR amplification using the sequence specific nucleic acid probe.
- the sample for use in the methods contains one or more tumor cells from the subject. In preferred embodiments, the sample for use in the methods contains circulating tumor DNA (ctDNA). In preferred embodiments, the sample is a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate.
- ctDNA tumor DNA
- the nucleic acids used in the method is isolated from a tumor cell sample from the subject.
- the nucleic acids used in the method are isolated from a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate from the subject.
- the nucleic acids for use in the assay is genomic DNA.
- the subject is diagnosed with or at risk of a recurrent or refractory form of a hyperproliferative disease, which subject has previously been treated with an irreversible BTK inhibitor.
- At risk of a recurrent or refractory form of a hyperproliferative disease in the context of this invention is defined as the subject having a mutation in a gene that results in expression of a mutated BTK protein that is unable to bind a BTK inhibitor, preferably an irreversible BTK inhibitor.
- the subject is diagnosed with recurrent or refractory form of a hyperproliferative disease, preferably a B-cell haematological malignancy that is resistant, or non-responsive to therapy with an irreversible BTK inhibitor.
- a hyperproliferative disease preferably a B-cell haematological malignancy that is resistant, or non-responsive to therapy with an irreversible BTK inhibitor.
- the subject is identified as having a mutation in a gene that results in expression of a mutated BTK protein that is unable to bind a BTK inhibitor, preferably an irreversible BTK inhibitor, which provides a risk of a recurrent or refractory form of a hyperproliferative disease, preferably a B-cell haematological malignancy that is resistant, or non-responsive to therapy with an irreversible BTK inhibitor.
- a BTK inhibitor preferably an irreversible BTK inhibitor, which provides a risk of a recurrent or refractory form of a hyperproliferative disease, preferably a B-cell haematological malignancy that is resistant, or non-responsive to therapy with an irreversible BTK inhibitor.
- the combination therapy is started when the B-cell haematological malignancy of the subject is relapsed or refractory, preferably wherein CLL/SLL is relapsed or refractory.
- the combination therapy is started when the B-cell haematological malignancy of the subject is relapsed or refractory and the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 , of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is C481 S.
- the pharmaceutical composition comprises a combination of an irreversible BTK inhibitor and a reversible BTK inhibitor, wherein the reversible BTK inhibitor is a compound according to any one of Formula (l-a) to (l-h), in a particular example a compound according to Formula A.
- Pharmaceutical compositions in accordance with the present invention may comprise, as one of the active ingredients (‘API’), compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt, hydrate or solvate thereof.
- composition refers to a composition comprising a compound according to the invention or a salt or solvate thereof and, as the case may be, one or more additional, non-toxic ingredients, which composition is in a form suitable for administration to a (human) subject, through any route of administration, and which composition is physiologically tolerated upon such administration.
- Exemplary, non-limiting unit dosage forms include a tablet (e.g., a chewable tablet), caplet, capsule (e.g., a hard capsule or a soft capsule), lozenge, film, strip, gelcap as well as any metered volume of a solution, suspension, syrup or elixir orthe like, which may be contained, for instance in a vial, syringe, applicator device, sachet, spray, micropump etc.
- the unit dosage form is a unit dosage form that is suitable for oral administration. Most preferably, it is a solid unit dosage form, such as a tablet.
- Pharmaceutically acceptable salts of compounds of the invention include the acid addition and base salts thereof, such as preferably the calcium, potassium or sodium salts.
- suitable salts reference is made “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
- salts of compounds according to the invention may be readily prepared by mixing together solutions of compounds according to the invention and the desired acid or base, as appropriate.
- the salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
- the degree of ionisation in the salt may vary from completely ionised to almost non-ionised.
- the compounds and the pharmaceutical compositions of the present invention are useful as inhibitors of tyrosine kinases, in particular of BTK.
- compounds of this invention are useful as inhibitors of tyrosine kinases that are important in hyper-proliferative diseases, especially in cancer, such as a B-cell hematological malignancy, and in the process of angiogenesis.
- the reversible BTK inhibitor compounds according to the invention having any one of Formula (l-a) to (l-h) and pharmaceutical compositions comprising these, can be used to treat or prevent a variety of conditions, diseases or disorders mediated by Bruton’s Tyrosine kinase (BTK).
- BTK Tyrosine kinase
- Such conditions, diseases or disorders include: cancers or tumors, including alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer including mast cell tumor and squamous cell carcinoma, breast and mammary cancer, ovarian cancer, prostate cancer, B cell hematological malignancy, lymphoma and leukemia (including but not limited to acute myelogenous leukemia, chronic myelogenous leukemia, mantle cell lymphoma, NHL B cell lymphomas (e.g.
- B-ALL marginal zone B-cell lymphoma, chronic lymphocytic leukemia, diffuse large B cell lymphoma, Burkitt lymphoma, mediastinal large B-cell lymphoma), Hodgkin lymphoma, NK and T cell lymphomas, TEL-Syk and ITK-Syk fusion driven tumors, myelomas including multiple myeloma, myeloproliferative disorders kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer, brain cancer, melanoma including oral and metastatic melanoma, Kaposi’s sarcoma, proliferative diabetic retinopathy, and angiogenic- associated disorders including solid tumors, and pancreatic cancer.
- the hyperproliferative disease is a B-cell hematological malignancy.
- the B-cell hematological malignancy is any one of chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), diffuse large B-cell lymphoma (DLBCL), activated B-cell diffuse large B-cell lymphoma (ABC-DLBCL), germinal center diffuse large B-cell lymphoma (GCB DLBCL), primary mediastinal B-cell lymphoma (PMBL), nonHodgkin lymphoma, Burkitt’s lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, precursor B-cell acute lymphoblastic leukemia, hairy cell leukemia, mantle cell lymphoma, B-cell prolymphocytic leukemia, lym- phoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma
- CLL
- the B-cell hematological malignancy is mantle cell lymphoma (MCL), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), diffuse large B- cell lymphoma (DLBCL), Waldenstrom macroglobulinemia (WM), follicular lymphoma (FL) and marginal zone lymphoma (MZL).
- MCL mantle cell lymphoma
- CLL chronic lymphocytic leukemia
- SLL small lymphocytic lymphoma
- DLBCL diffuse large B- cell lymphoma
- WM Waldenstrom macroglobulinemia
- FL follicular lymphoma
- MZL marginal zone lymphoma
- the B-cell malignancy is Mantle cell lymphoma (MCL) or chronic lymphocytic leukemia (CLL).
- MCL Mantle cell lymphoma
- CLL chronic lymphocytic leukemia
- compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
- compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
- the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks’s solution, Ringer’s solution, or physiological saline buffer.
- physiologically compatible buffers such as Hanks’s solution, Ringer’s solution, or physiological saline buffer.
- penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
- the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
- Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
- compositions for oral use can be obtained by combining the active compound with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
- suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
- disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
- Dragee cores are provided with suitable coatings.
- suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
- Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
- compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
- the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
- the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
- stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
- compositions may take the form of tablets or lozenges formulated in conventional manner.
- the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
- a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
- the dosage unit may be determined by providing a valve to deliver a metered amount.
- Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
- the compounds can be formulated for parenteral administration by injection, e.g. bolus injection or continuous infusion.
- Formulations for injection may be presented in unit dosage form, e.g. in ampoules or in multi-dose containers, with an added preservative.
- the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
- compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
- the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
- a suitable vehicle e.g., sterile pyrogen-free water
- the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
- the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly or by intramuscular injection).
- the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
- This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
- the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
- identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
- hydrophobic pharmaceutical compounds may be employed.
- Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
- Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
- the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
- sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
- additional strategies for protein stabilization may be employed.
- compositions also may comprise suitable solid or gel phase carriers or excipients.
- suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
- salts may be provided as salts with pharmaceutically compatible counterions.
- Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms.
- the compounds of the present invention can be prepared by methods well known in the art of organic chemistry. See, for example, J. March, ‘Advanced Organic Chemistry’ 4 th Edition, John Wiley and Sons. During synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This is achieved by means of conventional protecting groups, such as those described in T.W. Greene and P.G.M. Wutts ‘Protective Groups in Organic Synthesis’ 3 rd Edition, John Wiley and Sons, 1999. The protective groups are optionally removed at a convenient subsequent stage using methods well known in the art.
- Methyl (1 R,3/?)-3-aminocyclohexanecarboxylate hydrochloride (1.06 g, 5.47 mmol) was suspended in 10 mL water.
- Sodium bicarbonate (1.38 g, 16.4 mmol) in 10 mL water was added followed by a drop-wise addition of a solution A/-(benzyloxycarbonyloxy)succinimide (1 .50 g, 6.01 mmol) in dioxane (30 mL).
- the reaction mixture was stirred at room temperature o/n.
- the mixture was diluted with ethyl acetate (50 mL) and water (50 mL) and the bi-phasic system was stirred 30 minutes at room temperature.
- Triethylamine (10.4 mL, 74.62 mmol), 4-dimethylaminopyridine (605 mg, 4.95 mmol) di- fe/Y-butyl dicarbonate (13.5 g, 61.86 mmol) were added sequentially to a solution of 4- nitrobenzene sulfonamide (10 g, 49.46 mmol) in dichloromethane (100 mL).
- the reaction mixture was stirred for 30 minutes at room temperature.
- hydrochloric acid (1 N aqueous solution) until it becomes acidic.
- the organic layer was separated and washed with saturated sodium chloride aqueous solution, dried over sodium sulfate, filtered and then concentrated under reduced pressure.
- Example A Compound A
- Example A Compound A
- Lithium (E)-8-[3-[(1 R,3/?)-3-aminocyclohexyl]-1 -bromo-8-[(2,4-dimethoxyphenyl)- methylamino]imidazo[1 ,5-a]pyrazin-5-yl]oct-7-enoate (9.14 g, 15.07 mmol) was suspended dissolved in DMF (520 mL) and A/-ethylmorpholine (3.83 mL, 30.14 mmol) was added.
- Compound A as described herein is the same compound as compound having subformula 184 of co-pending PCT application PCT/EP2022/085765. Said compound having subformula 184 is also incorporated by reference.
- TMD8 diffuse large B-cell lymphoma cells were purchased from Tokyo Medical and Dental University. TMD8 cells expressing mutant BTK were created at Synthego Corporation. TMD8 cell lines expressing BTK C481 S, BTK T474I, BTK T474I/C481S, BTK V416L and BTK L528W were generated via CRISPR/Cas9. BTK T474I/C481 S expressing cells were generated by re- editing BTK C481 S cells. TMD8 mutant cell pools were enriched to obtain a 100% mutant genotype. The mutation status of all cell lines was confirmed via sequencing. Cells were cultured in MEM-alpha cell culture medium (cat. no. 22571038, Life Technologies), supplemented with 10% (v/v) heat-inactivated fetal bovine calf serum (Avantor, cat. no. 97068085) and 1 % penicillin/streptomycin.
- Inhibition of proliferation in response to compound was measured using the ATPIite 1 StepTM assay (cat. no. 6016736, Perkin Elmer).
- Percentage viability was used as the main y-axis signal.
- IC50S were fitted by non-linear regression using IDBS XLfit TM 5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC50.
- efficacy, GI50 and LD50 were also determined.
- WT-BTK, BTK C481 S, BTK T474I, BTK T474I/C481 S, BTK V416L and BTK L528W TMD8 cell lines were seeded at 800 cells per well in a white 384-well culture plate (cat. no. 781080, Greiner Bio-One) and allowed to rest for at least 4 hours at 37 °C, 95 % humidity, and 5 % CO2.
- Compound A pirtobrutinib, ibrutinib, acalabrutinib and nemtabrutinb were diluted in 3.16-fold dilution steps in 100% DMSO and further diluted in 20 mM Hepes, generating a dose response range from 316 nM to 0.0316 nM in the assay.
- DMSO was used as a vehicle control.
- Luminescence was recorded at 96 hours.
- Binding kinetics measurements on wt-BTK, BTK C481 S, BTK T474I (Surface Plasmon Resonance) Streptavidin-coated chips (Cat. No. BR100531), disposables and maintenance kits for Biacore were purchased from Cytiva (Eindhoven, The Netherlands). Biotinylated BTK WT enzym (Carna Biosciences, cat. no. 08-480-20N), BTK C481 S (Carna Biosciences, cat. no. OS- 417-20N), BTK T474I (Carna Biosciences, cat. no.
- the kinetic constants of the compounds were determined with single cycle kinetics with five consecutive injections with an increasing 15 compound concentration with ranges of 3.16 - 316 nM. Experiments were performed with an association time of 100 s per concentration and a dissociation time of 1200 s, except for compounds with a long target residence time, such as irreversible inhibitors, where dissociation time was increased. To circumvent problems of mass transport limitation, a flow rate of 30 pl/min was used. A blank run with the same conditions was performed before the compound was 20 injected. The SPR sensorgrams were analyzed with Biacore Evaluation Software by using a method of double referencing. First the reference channel was subtracted from the channel containing immobilized protein.
- pEF6/V5-HisB ThermoScientific cat. no. V96120 expression vectors containing either full-length wt-BTK (canonical sequence NM_000061 ), BTK C481S, BTK T474I, BTK T474I/C481S and BTK M437R (BaseClear) using LipofectamineTM 3000 (Invitrogen). Because the stop codon at the end of the coding sequence of BTK has been conserved, the His-tag present in the expression vector is not being used. Immediately after transfection, cells were cultured in medium without Geneticin, and after 24 hours 50 pg/ml geneticin was added.
- Cell based BTK inhibition wt-BTK, BTK C481S, BTK T474I, and BTK T474I/C481 S and BTK M437R expressing 293 cells were growing exponentially when seeded.
- Cells were seeded in culture medium without Blasticidin, at a density of 30.000 cells/well in 96-well plates (90 pL/well) (Greiner Bio-one, cat. no. 655182), and placed in a 37 °C CO 2 -incubator. Between 16 and 24 hours after seeding, 20 mM Hepes with compound or DMSO was added to the cells (10 pL).
- WT-BTK, BTK C481S, BTK T474I, BTK T474I/C481S and BTK M437R expressing 293 cells were growing exponentially when seeded, in culture medium, without blasticidin, at a density of 2.000.000 cells/4mL, in 25 cm 2 cell culture flasks (Greiner Bio-one, cat. no. 690175). Flasks were placed in a 37 °C CO 2 -incubator for 16-24 hours, followed by addition of compound (in medium). Two hours after addition of the inhibitors (Oh) cells are either collected or washed twice with 5 mL culture medium.
- Table 3 Proliferation data for BTK T474I TMD8 cells Proliferation assay of single compounds (monotherapy) in BTK T474I/C481S TMD8 cells.
- Table 4 Proliferation data for BTK T474I/C481 S TMD8 cells Proliferation assay of single compounds (monotherapy) in BTK V416L TMD8 cells. ( Figure 6)
- Table 7 Proliferation data for compounds in a mixture of BTK C481 S TMD8 cells + BTK V416L TMD8 cells.
- Table 9 KD and Target residence times (T (h)) for BTK inhibitors on wt-BTK, BTK C481S, BTK
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Hematology (AREA)
- Epidemiology (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Urology & Nephrology (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Oncology (AREA)
- Biotechnology (AREA)
- Biochemistry (AREA)
- Analytical Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Microbiology (AREA)
- Cell Biology (AREA)
- Hospice & Palliative Care (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to a medical use of a macrocyclic reversible BTK inhibitor and relates to a macrocyclic reversible BTK inhibitor for use in a method for treating a subject having a hyperproliferative disease. In particular embodiments, the subject receives or has received a Bruton's tyrosine kinase (BTK) inhibitor, preferably an irreversible BTK inhibitor, for treatment of the hyperproliferative disease. In particular embodiments, the method comprises a step of monitoring the subject over the course of therapy to determine whether the subject has a mutation in an endogenous gene encoding BTK.
Description
Medical use of a macrocyclic reversible BTK inhibitor
Field of the invention
The present invention relates to a medical use of a macrocyclic reversible BTK inhibitor and relates to a macrocyclic reversible BTK inhibitor for use in a method for treating a subject having a hyperproliferative disease. In particular embodiments, the subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor, preferably an irreversible BTK inhibitor, for treatment of the hyperproliferative disease. In particular embodiments, the method comprises a step of monitoring the subject over the course of therapy to determine whether the subject has a mutation in an endogenous gene encoding BTK.
Background of the invention
Kinases are enzymes that transfer a phosphate group from ATP to a protein. Kinases play an important role in regulating cellular functions such as cell proliferation, subcellular translocation, apoptosis, inflammation and metabolism (Attwood M.M. et al (2021) Nat Rev Drug Discov). The human kinome is composed of over 500 kinases. The development of smallmolecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy.
Bruton’s tyrosine kinase (BTK) is a member of the Src-related Tec family of protein kinases constituting a large subset of kinases, which play a central role in the regulation of a wide variety of cellular signaling processes. BTK plays a key role in B-cell receptor (BCR) signaling and a critical role in the regulation of survival, proliferation, activation and differentiation of B-lineage cells. Targeting of BTK with small molecule inhibitors such as the FDA approved covalent, irreversible, BTK inhibitors ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib has proven to be efficacious in several B-cell malignancies including Chronic Lymphocytic Leukemia (CLL), Mantle Cell Lymphoma (MCL), Waldenstrom’s Macroglobulinemia (WM) and Small Lymphocytic Lymphoma SLL.
BTK is also expressed and plays also pro-tumorigenic roles in several solid tumors (Xianhui Wang et al. 2021). In prostate cancer cells BTK inhibition with ibrutinib or acalabrutinib inhibited cell growth (Kokabee et al 2015). Ibrutinib has also been shown to inhibit in vivo (xenograft) breast cancer cell growth (Wang et al., 2016) and inhibition of BTK with ibrutinib blocked gastric cancer cell growth (Wang et al., 2016). BTK inhibitors have also showed inhibition of cellular proliferation and migration, and induced apoptosis and autophagy in glioblastoma cell lines (Wei et al., 2016; Wang et al., 2017).
Where tumors have a strong oncogenic drive from one particular kinase, specific amino acid mutations have been observed as acquired resistance mechanisms of targeted drug molecules. These mutations may naturally occur in very low levels but become more prevalent
upon drug treatment (pre-existing mutations), or these can also be produced by random mutation within tumors (acquired mutations) (Barouch-Bentov et al. 2011). Where such mutations have a clear advantage upon drug treatment (/.e. a mutation where the drug is no longer able to bind to the target) then resistance may rapidly develop and patients can relapse quickly. In these cases, the understanding of the impact and mechanism of these mutations has enabled additional drug discovery efforts to develop subsequent “generations” of drug molecules to target such resistance forms.
A drawback of the first generation BTK inhibitor, ibrutinib, is that drug resistance in B- cell malignancies can develop when BTK acquires mutations at the cysteine at position C481 of the kinase domain. This mutation abrogates the covalent binding of ibrutinib hampering its efficacy. Quinquenel et al. performed a ‘snapshot’ screening to determine the prevalence of resistance mutations and found that the presence of the BTK mutation was significantly associated with subsequent CLL progression. The correlation between disease progression, and the emergence and temporal dynamics of the most common resistance inducing C481 S BTK mutation have been determined for CLL patients receiving single-agent Ibrutinib treatment (Bodor et al. British Journal of Haematology 2021), Using digital droplet PCR the authors have been able to show that in 72.7% of the patients emergence of the BTK C481 S mutation preceded the symptoms of clinical relapse with a median of nine months. Woyach et al. showed that that detection of the C481 mutation in patients being treated with covalent, irreversible, BTK inhibitors is an indication for imminent relapse, usually within 12-18 months after detection (Woyach et al. (2017) J. Clin. Oncol. 35, 1437-1443). Since ibrutinib resistance confers poor survival, early detection of resistance provides clinically relevant information for the transition of affected patients to alternative treatment strategies.
Second-generation covalent, irreversible, BTK inhibitors, which include acalabrutinib, zanubrutinib, and tirabrutinib, offer greater BTK selectivity and therefore limited off-target toxicity. These inhibitors, however, do not overcome resistance by C481 mutation. To treat patients with relapsed CLL having C481 BTK mutations non-covalent, reversible, BTK inhibitors have been developed including LOXO-305 (pirtobrutinib) and ARQ-531 (nemtabrutinib) and vecabrutinib. These agents do not require covalent binding to the BTK C481 residue and effectively inhibit both wild type and mutant BTK with C481 substitutions. In preclinical studies, non-covalent BTK inhibitors including pirtobrutinib (LOXO-305), ARQ-351 and vecabrutinib, inhibited B-cell- receptor signaling in BTK C481 -mutant cell and animal models. Moreover, the phase 1-2 clinical trial of pirtobrutinib showed promising efficacy for patients with B-cell cancer who had previously been treated with covalent BTK inhibitors (with 62% of patients with CLL having a response), including patients with or without BTK C481 mutations (with a response occurring in 71 % and 66% of the patients, respectively) (Wang et al. (2022) N. Engl. J. Med. 386, 735-43). Pirtobrutinib was first disclosed in WO2017/103611 , ARQ-531 was disclosed in WO2017/111787 and vecabrutinib is disclosed in WO2013/185084. Further reversible BTK inhibitors are disclosed in WO2017/046604, , WQ2020/015735, WQ2020/239124, WQ2021/093839, WQ2020/043638,
WO2013/067274, WO2018097234, WO2013/010380, W02016/161570, WO2016/161571 , WO2016/106624, WO2016/106625, WO2016/106626, WO2016106623, WO2016/106628 and WO2016/109222.
Despite the successful treatment of patients with a C481 mutation with pirtobrutinib new acquired mutations have emerged after treatment with this non-covalent inhibitor. Mutations were identified in these patients that were clustered in the kinase domain of BTK (V416L, A428D, M437R, T474I, and L528W) and that conferred resistance to pirtobrutinib and other non- covalent, reversible, inhibitors.
The most well-known clinical documented BTK mutations for ibrutinib, acalabrutinib, zanubrutinib and pirtobrutinib are given in Figure 1 mutants (Nakoda et al. 2023).
Summary of the invention
In a first aspect of the invention, there is provided a method for treating a subject diagnosed with or at risk of a recurrent or refractory form of a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has previously been treated with a BTK inhibitor, preferably an irreversible BTK inhibitor, and wherein the method comprises administering to said subject an effective amount of said reversible BTK inhibitor, which is a compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is selected from the group consisting of:
W is an aryl group having 6-10 carbon or a heteroaryl group having 1-5 carbon; wherein any said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;
V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R1v)-NH-C(O)-, -CH(R1v)- ;
R1v is hydrogen or (1 -2C)alkyl;
U is an aryl group having 6-10 carbon or an heteroaryl group having 1 -5 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl, alkoxy, cycloalkyl and heterocycloalkyl group is optionally and independently substituted with one, two or three halogen; wherein R2 is of Formula (I l-a) to (ll-f) selected from the group consisting of:
(Formula ll-d) (Formula ll-e) (Formula ll-f) wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3- 6C)heterocycloalkyl, wherein Xi, X2 and X3 are independently selected from CH2, -CH2CH2-, O, N and a direct bond; wherein any of the cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with one or more substituents selected from halogen, hydroxy, (1 - 3C)alkyl, (1-3C)alkoxy, (1 -4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R3 and R4 together represent a linker having Formula (111-1 to HI-40) selected from the group consisting of:
whereby the ”” * marks the position of R3 in any one of Formula l-a to l-h, and whereby the 'I marks the position of R4 in any one of Formula I l-a to I l-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CDs, (1 -4C)alkyl, (1-5C)alkoxy, (3- 6C)cycloalkyl, (3-6C)cycloalkoxy and (1 -6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.
In another aspect of the invention, there is provided a method for treating a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, the method comprising the steps:
(a) monitoring the subject, preferably at predetermined intervals of time, over the course of the therapy for treatment of the hyperproliferative disease to determine whether the subject has a mutation in an endogenous gene encoding BTK that results in resistance or provides a risk of resistance of the subject to the therapy, preferably resistance of the subject to the irreversible BTK inhibitor, preferably the subject has a mutation in an endogenous gene encoding BTK that results in expression of a BTK protein with a modification at an amino acid position corresponding to amino acid position 481 of the amino acid sequence according to SEQ ID NO: I; and
(b) if the subject has shown said mutation, administering to the subject a reversible BTK inhibitor, in particular if the subject has the BTK modification corresponding to amino acid position 481 identified in step (a), wherein the reversible BTK inhibitor is a compound according to the invention having any one of Formula (l-a) to (l-h).
In another aspect of the invention, there is provided a method for treating a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, wherein the method comprises: optionally stopping the administration of the irreversible BTK inhibitor, and administering to said subject an effective amount of a reversible BTK inhibitor, wherein the reversible BTK inhibitor is a compound according to the invention having any one of Formula (I- a) to (l-h).
In other aspects of the invention, there is provided a reversible BTK inhibitor for use in a method according to the invention which is a compound of any one of Formula (l-a) to (l-h).
In other aspects of the invention, there is provided use of a reversible BTK inhibitor according to the invention in the manufacture of a medicament for treating a subject having a hyperproliferative disease, preferably treating a subject having a B-cell hematological malignancy.
The present inventors have surprisingly found that the reversible BTK inhibitor according to the invention is very effective in inhibiting BTK mutants, e.g. in inhibiting B-cell lymphoma cells comprising two separate cell pools each containing a different mutation in BTK. Additionally, the inventors have found that the reversible BTK inhibitor according to the invention is very effective in treating subjects having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor.
The present invention will be illustrated further by means of the following non-limiting examples.
Definitions
The term "pharmaceutical composition” as used herein has its conventional meaning and refers to a composition which is pharmaceutically acceptable.
The term "pharmaceutically acceptable” as used herein has its conventional meaning and refers to compounds, material, compositions and/or dosage forms, which are, within the scope of sound medical judgment suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
The term "effective amount" as used herein, refers to an amount of the compound of the invention, and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventive effect when administered to a subject suffering from a BTK-mediated disease or disorder. In the combination therapies of the present invention, as effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount.
A "subject" is a human or non-human mammal. In one embodiment, a subject is a human. The term "controlling” is intended to refer to all processes wherein there may be a slowing, interrupting, arresting or stopping of the progression of the diseases and conditions affecting the mammal. However, "controlling” does not necessarily indicate a total elimination of all disease and condition symptoms, and is intended to include prophylactic treatment.
The term "excipient” as used herein has its conventional meaning and refers to a pharmaceutically acceptable ingredient, which is commonly used in the pharmaceutical technology for preparing a granulate, solid or liquid oral dosage formulation.
The term "salt” as used herein has its conventional meaning and includes the acid addition and base salts of the compound of the invention.
The term "solvate” as used herein has its conventional meaning. One or more compounds of the invention or the pharmaceutically acceptable salts thereof may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. "Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association Involves varying degrees of ionic and covalent bonding. Including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H2O and includes any hydrate of the compound or the salt of said compound.
The term "treatment” as used herein has its conventional meaning and refers to curative, disease controlling, palliative and prophylactic treatment.
The term "unit dosage form” has its conventional meaning and refers to a dosage form which has the capacity of being administered to a subject, preferably a human, to be effective, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose comprising the therapeutic agent, i.e. the compound of the invention.
The term "BTK” as used herein has its conventional meaning and refers to Bruton's Tyrosine Kinase. Bruton's tyrosine kinase (BTK) is a member of the Src-related Tec family of protein kinases which are a large subset of kinases which play a central role in the regulation of a wide variety of cellular signaling processes. BTK plays a key role in the B-cell receptor signaling and a critical role in the regulation of survival, proliferation, activation and differentiation of B- lineage cells. Targeting of BTK with small molecule inhibitors such as the FDA approved irreversible BTK inhibitors ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib has proven to be efficacious in several B-cell malignancies including Chronic Lymphocytic Leukemia (CLL), Mantle Cell Lymphoma (MCL), Waldenstrom’s Macroglobulinemia (WM) and Small Lymphocytic Lymphoma SLL. Combinations of BTK inhibitors with other novel drugs or regimens results in more profound responses and much higher rates of minimal residual disease negativity.
The term "BTK inhibitor” as used herein has its conventional meaning and refers to an inhibitor for BTK. A BTK inhibitor may be a small molecule inhibitor. Inhibitors may be irreversible inhibitors, such as by forming a covalent bond, and may be reversible inhibitors, which may form a reversible interaction with BTK.
The term “covalent BTK inhibitor” as used herein has its conventional meaning and refers to a BTK inhibitor that reacts with its target protein (BTK) to form a covalent complex in which the protein has lost its function. Covalent inhibitors can be reversible or irreversible, depending on the rate of the reverse reaction. The terms 'covalent inhibitor' and 'irreversible inhibitor' are often used interchangeably.
The term "irreversible BTK inhibitor” as used herein has its conventional meaning and refers to a BTK inhibitor that possesses an off-rate that is slow relative to the rate of re-synthesis of the target protein (BTK) in vivo, so that once the target protein is inhibited, it does not regain activity.
The term “rate of re-synthesis” as used herein has its conventional meaning and refers to the rate at which a cell and/or organism replaces a protein target with freshly synthesized functional protein. The re-synthesis rate defines the rate at which an irreversibly inhibited protein target will recover activity in vivo, once the inhibitor is no longer present.
The term "reversible BTK inhibitor” as used herein has its conventional meaning and refers to a BTK inhibitor that inactivates the BTK enzyme through non-covalent, transcient, interactions. Unlike an irreversible inhibitor, a reversible inhibitor can dissociate from the enzyme.
The term "mutant-BTK” as used herein has its conventional meaning and refers to mutations of BTK. Mutations of BTK may be referred to by an altered or modified amino acid
target (such as C as single-letter amino acid code for cysteine) at a certain position of the BTK structure (such as 481). Additionally, the amino acid substitution at the modification position may be referred to by an additional amino acid single-letter amino acid code, such as C481 S for serine substitution and C481T for threonine substitution of cysteine at the 481 position.
The term “modification" as used herein has its conventional meaning and refers to modification of a sequence of amino acids of a polypeptide, protein or a sequence of nucleotides in a nucleic acid molecule and includes deletions, insertions, and replacements of amino acids and nucleotides, respectively.
The term "resistance to BTK inhibitor” as used herein has its conventional meaning and refers to a BTK inhibitor, which shows a reduction in effectiveness, after being effective initially in treating a hyperproliferative disease, in particular a B-cell hematological malignancy.
A drawback of the currently approved irreversible inhibitors is that patients treated with these inhibitors can develop drug resistance when BTK proteins with variations at the catalytic site are not able to bind efficiently to irreversible inhibitors in patients. This is a rather common event in patients treated with irreversible inhibitors and who experience relapse. A major mechanism for the acquired resistance is the emergence of BTK cysteine 481 (C481) mutations. These mutations hamper binding of irreversible inhibitors such as ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib which form a covalent bond with this amino acid. Other mutations / modifications that can result in acquired resistance of irreversible covalent and/or reversible non- covalent BTK inhibitors are valine 416 (V416), alanine 428 (A428), methionine 437 (M437), threonine 474 (T474) and leucine 528 (L528) modifications, which can reduce BTK inhibitor binding to BTK.
By the term “drug resistance" as used herein is meant the major cause of cancer treatment failure. While a treatment may be effective initially, the heterogeneity of cancer and its ability to adapt can allow the cancer to become resistant to the treatment and regrow.
The term "relapse” as used herein has the conventional meaning and refers to evidence of disease progression in a patient who has previously achieved criteria of a complete response or partial remission.
The term ‘’disease progression” as used herein has the conventional meaning and refers to a measured increase in tumor size or tumor burden.
The term “ recurrent or refractory form of a hyperproliferative disease” as used herein has the conventional meaning and refers to a particular cancer that is resistant, or non-responsive to therapy with a particular therapeutic agent. A cancer can be refractory to therapy with a particular therapeutic agent either from the onset of treatment (i.e., non-responsive to initial exposure to the therapeutic agent), or as a result of developing resistance to the therapeutic agent, either over the course of a first treatment period with the therapeutic agent or during a subsequent treatment period with the therapeutic agent.
The term ‘’acquired drug resistance" as used herein has the conventional meaning and refers to resistance of a drug caused by mutations in the target protein. The mutations in the target protein hamper the binding of drug resulting in a regrowth of the tumor
The term ‘’clinical drug resistance” as used herein has the conventional meaning and refers to growth of a tumour while the patient is on treatment, that develops following after an initial clinical benefit (a clinical response or prolonged stable disease).
The term "wt-BTK” or “WT-BTK” or “BTKWT” as used herein has its conventional meaning and refers to wild-type Bruton’s Tyrosine Kinase. A wild-type BTK has the regular meaning of a phenotype of the typical form of BTK as it occurs in nature. Originally, the wild-type was conceptualized as a product of the standard "normal" allele at a locus, in contrast to that produced by a non-standard, "mutant" allele.
The term "macrocycle” as used herein has its conventional meaning and refers to a part of a molecule containing a ring consisting of 12 or more ring atoms forming said ring. In an example, a twelve membered ring consist of 12 atoms forming said ring.
The term "binding affinity (KD)” as used herein has its conventional meaning and refers to the equilibrium dissociation constant which is an inverse measure of the affinity of a proteinligand (small molecule) pair under equilibrium conditions. The value of KD is mathematically equivalent to the ratio kotr/kon (or kd/ka) measured using Surface Plasmon Resonance (SPR).
The term "association rate constant” or “on-rate (kon or ka)” as used herein has its conventional meaning and refers to a second-order rate constant that quantifies the rate at which a free ligand and free protein combine (through collisional encounters) to form a binary proteinligand complex.
The term "dissociation rate constant” or “off-rate (kOft or kd)” as used herein has its conventional meaning and refers to a first-order rate constant that quantifies the rate at which a binary protein-ligand complex dissociates to the free ligand and free protein.
The term "target residence time tau (r)” as used herein has its conventional meaning and refers to the time a compound resides on its target. Target residence time (T) can be determined according to the method as described below in the experimental section.
The term “IC50” as used herein has its conventional meaning and refers to the concentration of a substance that results in a 50% effect on some measure of biochemical and/or cellular function or substance-target binding interaction.
The term “pICso” as used herein has its conventional meaning and refers to the negative logarithm of the IC50 in molar concentration.
The term “G/50” as used herein has its conventional meaning and refers to the concentration of a substance that inhibits cell growth by 50%.
The term “LD50” as used herein refers to the concentration of a substance that results in 50% cell death.
The term “efficacy” as used herein has its conventional meaning and refers to the effect that is reached at a particular concentration of a substance.
A bicyclic ringsystem, as used herein, refers to heterocyclic (heterocyclyl) groups, to cyclic groups having carbon groups only, i.e. without hetero atoms, within the cycle, and to combinations of a heterocyclic (heterocyclyl) group and a cyclic group having carbon groups only, i.e. without hetero atoms, within the cycle.
A monocylic ringsystem, as used herein, refers both to a heterocyclic (heterocyclyl) group, and to a cyclic group having carbon groups only, i.e. without hetero atoms, within the cycle.
A heterocyclic (heterocyclyl) group, as used herein, refers to both heteroaryl groups and heterocycloalkyl groups.
A heterobicyclic group, as used herein, refers to a bicyclic group having one or more heteroatoms, which is saturated, partially unsaturated or unsaturated.
As used herein, aromatic groups (or aryl groups) include aromatic carbocyclic ring systems (e.g. phenyl) and fused polycyclic aromatic ring systems (e.g. naphthyl and 1 , 2,3,4- tetrahydronaphthyl).
The term "alkyl," as used herein, refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond having the specified number of carbon atoms. In different embodiments, an alkyl group contains, for example, from 1 to 6 carbon atoms (1 - 6C)Alkyl or from 1 to 3 carbon atoms (1 -3C)Alkyl. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl. In one embodiment, an alkyl group is linear. In another embodiment, an alkyl group is branched.
Unless specified otherwise, "alkyl" includes both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbon atoms; for example, "(1-6C)Alkyl" includes all of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl. "Alkylene" refers to both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbons, and having two terminal end chain attachments; for example, the term "A-C4 alkylene-B" represents, for example, A-CH2-CH2-CH2-CH2-B, A-CH2- CH2-CH(CH3)-CH2-B, A-CH2-CH(CH2CH3)-B, A-CH2-C(CH3)(CH3)-B, and the like.
The term "alkylcarbonyl," as used herein, refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond attached to a carbonyl group, wherein the aliphatic hydrocarbon group has the specified number of carbon atoms. In different embodiments, an alkyl group or aliphatic hydrocarbon group contains, for example, from 1 to 6 carbon atoms (1 -6C)Alkyl or from 1 to 3 carbon atoms (1 -3C)Alkyl. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl. In one embodiment, an alkyl group is linear. In another embodiment, an alkyl group is branched.
Cycloalkyl means a cycloalkyl group having the recited number of carbon atoms, with the same meaning as previously defined, such as cyclopropyl, cyclobutyl, or cyclopentyl.
"Cycloalkyl" refers to a cycloalkyl-group represented by an indicated number of carbon atoms; for example "(3-6C)cycloalkyl" includes cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
Heterocycloalkyl means a cycloalkyl group having the recited number of carbon atoms, and 1-3 heteroatoms selected from N, O and/or S, with the same meaning as previously defined.
Haloalkyl means a branched or unbranched alkyl group having the recited number of carbon atoms, in which one and up to all hydrogen atoms are replaced by a halogen; halogen is as defined herein. Examples of such branched or straight chained haloalkyl groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl and n- butyl substituted independently with one or more halogens, e.g., fluoro, chloro, bromo and iodo.
For example, a halo(1 -3C)alkyl means a branched or unbranched alkyl group having 1 ,2, or 3 carbon atoms, in which at least one hydrogen atom is replaced by a halogen. Examples of "haloalkyl" include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 1 - fluoroethyl, 2-fluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, and perfluoro-n-propyl.
Alkoxy means an alkoxy group having the recited number of carbon atoms, the alkyl moiety having the same meaning as previously defined, e.g., "Alkoxy" refers to an alkyl-O-group represented by a linear or branched alkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "(1 -6C)Alkoxy" includes -OCH3, -O-CH2CH3, - OCH(CH3)2, -O(CH2)5CH3, and the like.
Cycloalkoxy means a cycloalkyl group having the recited number of carbon atoms, with the same meaning as previously defined, attached via a ring carbon atom to an exocyclic oxygen atom, such as cyclopropoxyl, cyclobutoxyl,or cyclopentoxyl. "Cycloalkoxy" refers to a cycloalkyl- O-group represented by a cycloalkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "(3-6C)cycloalkoxy" includes cyclopropyl-O-, cyclobutyl- O-, cyclopentyl-O-, or cyclohexyl-O-.
Heterocycloalkoxy means a cycloalkyl group having the recited number of carbon atoms, and 1-3 heteroatoms selected from N, O and/or S, with the same meaning as previously defined, attached via a ring carbon atom to an exocyclic oxygen atom.
Unless otherwise specifically noted as only "unsubstituted" or only "substituted", alkyl groups are unsubstituted or substituted with 1 to 3 substituents on each carbon atom.
It should be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
The terms first, second, third and the like in the description and in the claims, are used for distinguishing between for example similar elements, compositions, constituents in a composition, or separate method steps, and not necessarily for describing a sequential or chronological order. The terms are interchangeable under appropriate circumstances and the embodiments of the invention can operate in other sequences than described or illustrated herein, unless specified otherwise.
Furthermore, the various embodiments, although referred to as “preferred” or “e.g." or “for example” or “in particular” and the like are to be construed as exemplary manners in which the invention may be implemented rather than as limiting the scope of the invention.
The term “comprising”, used in the claims, should not be interpreted as being restricted to for example the elements or the method steps or the constituents of a compositions listed thereafter; it does not exclude other elements or method steps or constituents in a certain composition. It needs to be interpreted as specifying the presence of the stated features, integers, (method) steps or components as referred to, but does not preclude the presence or addition of one or more other features, integers, steps or components, or groups thereof. Thus, the scope of the expression “a method comprising steps A and B” should not be limited to a method consisting only of steps A and B, rather with respect to the present invention, the only enumerated steps of the method are A and B, and further the claim should be interpreted as including equivalents of those method steps. Thus, the scope of the expression “a composition comprising components A and B” should not be limited to a composition consisting only of components A and B, rather with respect to the present invention, the only enumerated components of the composition are A and B, and further the claim should be interpreted as including equivalents of those components.
In addition, reference to an element or a component by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element or component are present, unless the context clearly requires that there is one and only one of the elements or components. The indefinite article "a" or "an" thus usually means "at least one".
Legend to the figures
Figure 1 : Map of clinically documented BTK mutants.
Figure 2: Proliferation assay of single compounds (monotherapy) in wt-BTK TMD8 cells.
Figure 3: Proliferation assay of single compounds (monotherapy) in BTK C481 S TMD8 cells.
Figure 4: Proliferation assay of single compounds (monotherapy) in BTK T474I TMD8 cells.
Figure 5: Proliferation assay of single compounds (monotherapy) in BTK T474I/C481 S TMD8 cells.
Figure 6: Proliferation assay of single compounds (monotherapy) in BTK V416L TMD8 cells.
Figure 7: Proliferation assay of single compounds (monotherapy) in BTK L528W TMD8 cells.
Figure 8: Proliferation assay of single compounds (monotherapy) in a mixture of BTK
C481 S TMD8 cells and BTK V416L TMD8 cells.
Figure 9: Proliferation assay of single compounds (monotherapy) in a mixture of BTK C481 S TMD8 cells and BTK T474I TMD8 cells.
Figure 10A: Washout pirtobrutinib and compound A in wt-BTK GripTite 293 MSR cells.
Figure 10B: Quantification of washout pirtobrutinib and compound A in wt-BTK GripTite 293 MSR cells.
Figure 11 A: Washout pirtobrutinib and compound A in BTK C481 S GripTite 293 MSR cells.
Figure 11 B: Quantification of washout pirtobrutinib and compound A in BTK C481 S GripTite 293 MSR cells.
Figure 12A: Washout pirtobrutinib and compound A in BTK T474I GripTite 293 MSR cells.
Figure 12B: Quantification of washout pirtobrutinib and compound A in BTK T474I GripTite 293 MSR cells.
Figure 13A: Washout pirtobrutinib and compound A in BTK T474I/C481 S GripTite 293 MSR cells.
Figure 13B: Quantification of washout pirtobrutinib and compound A in BTK T474I/C481 S GripTite 293 MSR cells. Figure 14A: Washout pirtobrutinib and compound A in BTK M437R GripTite 293 MSR cells.
Figure 14B: Quantification of washout pirtobrutinib and compound A in BTK M437R GripTite 293 MSR cells.
Figure 15A: Western blot Dose Response Curve results of compound A in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
Figure 15B: Western blot Dose Response Curve results of pirtobrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
Figure 15C: Western blot Dose Response Curve results of ibrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
Figure 15D: Western blot Dose Response Curve results of acalabrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
Figure 15E: Western blot Dose Response Curve results of nemtabrutinib in wt-BTK, BTK C481 S, BTK T474I and BTK C481 S/T474I expressing 293 cells.
Figure 15F: Western blot Dose Response Curve results of ibrutinib, acalabrutinib, pirtobrutinib, nemtabrutinib and compound A in BTK M437R expressing 293 cells.
Figure 16: Ibrutinib resistance mutations can be detected before clinical relapse. For 20 patients with a detectable mutation in BTK or PLCG2 at time of relapse, samples before relapse were analyzed retrospectively to determine the interval of time between mutation detection and clinical relapse. An initial clone could be detected at an estimated median of 9.3 months before relapse. UPN, unique patient number (Woyach et al. 2017).
Detailed description of the invention
The present inventors have surprisingly established that the macrocyclic reversible BTK inhibitor according to the invention is very effective in inhibiting BTK mutants, e.g. in inhibiting B-
cell lymphoma cells comprising two separate cell pools each containing a different mutation in BTK. Additionally, the inventors have found that the reversible BTK inhibitor according to the invention is very effective in treating subjects having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor.
Surprisingly, the inventors have found that compounds according to the invention provide an improved reversible binding activity towards wild-type BTK and/or BTK mutants. The compounds according to the invention have any one of Formula (l-a) to (l-h), which contains a macrocyclic moiety, in combination with BTK specific pharmacophores (e.g. based on ligands for binding to BTK) to provide a binding activity towards wild-type BTK and/or BTK mutants through improved reversible binding. In particular it has been found that the compound provides a considerable longer residence time than what is typically obtained with reversible BTK inhibitors.
In preferred embodiments, the reversible BTK inhibition compounds according to the invention have any one of Formula (l-a) to (l-h), which are extensively described in co-pending PCT application PCT/EP2021/085641 and in PCT application PCT/EP2022/085765. The reversible BTK inhibitors according to the present invention include the (macrocyclic) BTK inhibition compounds of the co-pending PCT application PCT/EP2021/085641 and of the PCT application PCT/EP2022/085765 and are incorporated by reference, including the exemplary compounds having sub-formula 1 - 226 and the described syntheses routes for manufacturing the compounds having any one of Formula (l-a) to (l-h).
Embodiments
Reversible BTK inhibitors
In preferred embodiments the reversible BTK inhibitor is a compound according to Formula (l-a) or (l-b) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound comprises a bicyclic scaffold selected from:
-b)
Wherein R1 is any one of:
wherein R2w is selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any said alkyl or alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R3u is selected from hydrogen, halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3- 6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R2 is selected from the group consisting of:
(Formula ll-d) (Formula ll-f) (Formula Il-a5) (Formula Il-b4) wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with hydroxy, methyl, acetyl or methoxy; wherein R3 and R4 together represent a linker having Formula selected from the group consisting of:
whereby the T~ * marks the position of R3 in any one of Formula l-a to l-b, and whereby the
marks the position of R4 in any one of Formula I l-a to I l-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CD3, (1 -4C)alkyl, (1-5C)alkoxy, (3- 6C)cycloalkyl, (3-6C)cycloalkoxy and (1 -6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.
In particular embodiments the reversible BTK inhibitor is a compound according to Formula A:
(A), or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
Irreversible BTK inhibitors
In preferred embodiments, the irreversible BTK inhibitor, which the subject has received, is selected from the group consisting of: ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, spebrutinib, branebrutinib, evobrutinib, remibrutinib, tolebrutinib, orelabrutinib, elsubrutinib, edralbrutinib, ACP-5862, and pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
In a preferred embodiment, the irreversible BTK inhibitor is ibrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
In a preferred embodiment, the irreversible BTK inhibitor is acalabrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
In a preferred embodiment, the irreversible BTK inhibitor is zanubrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
In a preferred embodiment, the irreversible BTK inhibitor is tirabrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
BTK Protein Sequence
SEQ ID NO: 1 tyrosine-protein kinase BTK [Homo sapiens] 10 20 30 40 50 60
MAAVI LES I F LKRSQQKKKT S PLNFKKRLF LLTVHKLSYY EYDFERGRRG SKKGS IDVEK
70 80 90 100 110 120
ITCVETVVPE KNPPPERQI P RRGEESSEME QI S I IERFPY PFQVVYDEGP LYVFS PTEEL
130 140 150 160 170 180
RKRWIHQLKN VIRYNSDLVQ KYHPCFWIDG QYLCCSQTAK NAMGCQI LEN RNGSLKPGSS
190 200 210 220 230 240
HRKTKKPLPP TPEEDQI LKK PLPPE PAAAP VSTSELKKVV ALYDYMPMNA NDLQLRKGDE
250 260 270 280 290 300
YFI LEESNLP WWRARDKNGQ EGYI PSNYVT EAEDS IEMYE WYSKHMTRSQ AEQLLKQEGK
310 320 330 340 350 360
EGGFIVRDSS KAGKYTVSVF AKSTGDPQGV IRHYVVCSTP QSQYYLAEKH LFSTI PELIN
370 380 390 400 410 420
YHQHNSAGLI SRLKYPVSQQ NKNAPSTAGL GYGSWE IDPK DLTFLKELGT GQFGVVKYGK
430 440 450 460 470 480
WRGQYDVAIK MIKEGSMSED E FIEEAKVMM NLSHEKLVQL YGVCTKQRPI FI ITEYMANG
490 500 510 520 530 540
CLLNYLREMR HRFQTQQLLE MCKDVCEAME YLESKQFLHR DLAARNCLVN DQGVVKVSDF
550 560 570 580 590 600
GLSRYVLDDE YTSSVGSKFP VRWS PPEVLM YSKFSSKSDI WAFGVLMWE I YSLGKMPYER
610 620 630 640 650 660
FTNSETAEHI AQGLRLYRPH LASEKVYTIM YSCWHEKADE RPTFKI LLSN I LDVMDEES *
Mutant BTK
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the protein modification is C481 S.
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 of the amino acid sequence according to SEQ ID NO: I, wherein the modification is a substitution of cysteine to an amino acid selected from leucine, isoleucine, valine, alanine, glycine, methionine, serine, threonine, phenylalanine, tryptophan, lysine, arginine, histidine, proline, tyrosine, asparagine, glutamine, aspartic acid and glutamic acid at amino acid position 481 of the BTK protein.
In a more preferred embodiment, the modification is a substitution of cysteine to serine at amino acid position 481 of the BTK protein.
In a preferred embodiment, the subject is identified as having one or more mutations in a gene that results in expression of a BTK protein with a modification at one or more amino acid positions selected from the group consisting of: 416, 428, 437, 474, 481 and 528.
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 416 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is V416L.
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 428 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is A428D.
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 437 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is M437R.
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 474 of the amino acid sequence set according to SEQ ID NO: I, more preferably wherein the mutant modification is T474L
In a preferred embodiment, the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 528 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is L528W.
Prior therapy
In preferred embodiments, prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, ibrutinib before starting a new treatment option for managing the same condition.
In preferred embodiments, prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, acalabrutinib before starting a new treatment option for managing the same condition.
In preferred embodiments, prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease,
preferably an irreversible BTK inhibitor, more preferably, zanubrutinib before starting a new treatment option for managing the same condition.
In preferred embodiments, prior therapy has been given to a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, where the subject has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, tirabrutinib before starting a new treatment option for managing the same condition.
In preferred embodiments, the irreversible BTK inhibitor is administered during the prior therapy in an amount in the range of 70 - 750 mg / day and / or wherein the irreversible BTK inhibitor is administered using one or more unit doses having an amount in the range of 70 - 750 mg / unit dose.
In preferred embodiments, the irreversible BTK inhibitor is administered during the prior therapy daily at a dose selected from the group consisting of 70 mg, 100 mg, 140 mg, 160 mg, 200 mg, 280 mg, 320 mg, 420 mg, 480 mg, 560 mg per day.
In preferred embodiments, the irreversible BTK inhibitor is administered during the prior therapy twice daily at a dose of 80 mg, 100 mg or 160 mg..
Maintenance Therapy
In preferred embodiments, maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, ibrutinib.
In preferred embodiments, maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, acalabrutinib.ln preferred embodiments, maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, zanubrutinib.
In preferred embodiments, maintenance therapy is given to a subject having a hyperproliferative disease, preferably a B-cell haematological malignancy, where the subject has received and still is receiving a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, more preferably, tirabrutinib.
Provided herein are embodiments of methods including maintenance therapy of subject having a B-cell hematological malignancy. In some embodiments, the methods for maintenance therapy comprise treating a B-cell hematological malignancy with a covalent and/or irreversible BTK inhibitor for an initial treatment period, followed by a maintenance therapy regimen. In some embodiments, the methods for maintenance therapy comprise treating a B-cell haematological malignancy with a covalent and/or irreversible BTK inhibitor for a period of six months or longer, such as, for example, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, 13 months, 14 months, 15 months, 16 months, 17 months, 18 months, 19 months, 20 months, 21 months, 22 months, 23 months, 24 months, 25 months, 26 months, 27 months, 28 months, 29 months, 30 months, 31 months, 32 months, 33 months, 34 months, 35 months, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years or longer.
Monitoring subject
In preferred embodiments, the subject is monitored over the course of the therapy with a BTK inhibitor for treatment of the hyperproliferative disease to determine whether the subject has a mutation in an endogenous gene encoding BTK that results in or provides a risk of resistance of the subject to the therapy, preferably resistance of the subject to the irreversible BTK inhibitor.
In preferred embodiments, the subject is monitored at predetermined intervals of time over the course of the therapy for treatment of the hyperproliferative disease, wherein the predetermined interval of time is every week, every 2 weeks, every 3 weeks, every month, every 2 months, every 3 months, every 4 months, every 5 months, every 6 months, every 7 months, every 8 months, every 9 months, every 10 months, every 11 months, every year following the first administration of the covalent and/or irreversible BTK inhibitor. The treatment which is monitored is in particular a treatment of administration of an irreversible BTK inhibitor.
Methods for monitoring subject
In some embodiments of the methods, testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 481 of the BTK protein. In some embodiments, PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 481 of the BTK protein. In some embodiments, the method comprises seguencing the amplified nucleic acids. In some embodiments of the methods, testing comprises performing deep seguencing of nucleic acids that encode amino acid position 481 of the BTK protein.
In some embodiments of the methods, testing comprises contacting the nucleic acids with a seguence specific nucleic acid probe, wherein the seguence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 481 ;
and (b) does not bind to nucleic acids encoding the wild-type BTK having cysteine at amino acid position 481 . In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In some embodiments of the methods, testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 416 of the BTK protein. In some embodiments, PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 416 of the BTK protein. In some embodiments, the method comprises sequencing the amplified nucleic acids. In some embodiments of the methods, testing comprises performing deep sequencing of nucleic acids that encode amino acid position 416 of the BTK protein.
In some embodiments of the methods, testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 416; and (b) does not bind to nucleic acids encoding the wild-type BTK having valine at amino acid position 416. In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In some embodiments of the methods, testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 428 of the BTK protein. In some embodiments, PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 428 of the BTK protein. In some embodiments, the method comprises sequencing the amplified nucleic acids. In some embodiments of the methods, testing comprises performing deep sequencing of nucleic acids that encode amino acid position 428 of the BTK protein.
In some embodiments of the methods, testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 428; and (b) does not bind to nucleic acids encoding the wild-type BTK having alanine at amino acid position 428. In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In some embodiments of the methods, testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 437 of the BTK protein. In some embodiments, PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 437 of the BTK protein. In some embodiments, the method comprises sequencing the amplified nucleic acids. In some embodiments of the methods, testing comprises performing deep sequencing of nucleic acids that encode amino acid position 437 of the BTK protein.
In some embodiments of the methods, testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 437; and (b) does not bind to nucleic acids encoding the wild-type BTK having methionine at amino acid position 437. In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In some embodiments of the methods, testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 474 of the BTK protein. In some embodiments, PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 474 of the BTK protein. In some embodiments, the method comprises sequencing the amplified nucleic acids. In some embodiments of the methods, testing comprises performing deep sequencing of nucleic acids that encode amino acid position 474 of the BTK protein.
In some embodiments of the methods, testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 474; and (b) does not bind to nucleic acids encoding the wild-type BTK having threonine at amino acid position 474. In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In some embodiments of the methods, testing comprises performing polymerase chain reaction (PCR) amplification of nucleic acids that encode amino acid position 528 of the BTK protein. In some embodiments, PCR amplification comprises using a pair of oligonucleotide primers that flank the region encoding amino acid position 528 of the BTK protein. In some embodiments, the method comprises sequencing the amplified nucleic acids. In some embodiments of the methods, testing comprises performing deep sequencing of nucleic acids that encode amino acid position 528 of the BTK protein.
In some embodiments of the methods, testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 528; and (b) does not bind to nucleic acids encoding the wild-type BTK having tryptophane at amino acid position 528. In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In some embodiments, the sample for use in the methods contains one or more tumor cells from the subject. In some embodiments, the sample for use in the methods contains cell free DNA (cfDNA). In some embodiments, the sample for use in the methods contains circulating tumor DNA (ctDNA). In some embodiments, the sample for use in the methods contains tumor derived extracellular vesicles (EV). In some embodiments, the sample is a tumor biopsy sample,
a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate.
In some embodiments of the methods, the nucleic acids used in the method is isolated from a tumor cell sample from the subject. In some embodiments of the methods, the nucleic acids used in the method are isolated from extracellular vesicles (EV) isolated from the subject. In some embodiments of the method the nucleic acids used in the method are isolated from a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate from the subject.
In some embodiments of the methods, the nucleic acids for use in the assay are RNA or DNA. In some embodiments of the methods, the nucleic acids for use in the assay are genomic DNA. In some embodiments of the methods, the nucleic acids for use in the assay are total RNA. In some embodiments of the methods, the nucleic acids for use in the assay are messenger RNA (mRNA).
In some embodiments of the methods, the nucleic acids for use in the assay are complementary DNA (cDNA).
In some embodiments of the methods, the method further comprises isolating mRNA from the RNA sample. In some embodiments of the methods, the method further comprises reverse transcription of the total RNA, or mRNA, into cDNA.
In preferred embodiments of the methods, testing comprises contacting the nucleic acids with a sequence specific nucleic acid probe, wherein the sequence specific nucleic acid probe: (a) binds to nucleic acids encoding a modified BTK that is modified at amino acid position 481 ; and (b) does not bind to nucleic acids encoding the wild-type BTK having cysteine at amino acid position 481. In some embodiments of the methods, testing comprises PCR amplification using the sequence specific nucleic acid probe.
In preferred embodiments, the sample for use in the methods contains one or more tumor cells from the subject. In preferred embodiments, the sample for use in the methods contains circulating tumor DNA (ctDNA). In preferred embodiments, the sample is a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate.
In preferred embodiments of the methods, the nucleic acids used in the method is isolated from a tumor cell sample from the subject. In preferred embodiments of the method the nucleic acids used in the method are isolated from a tumor biopsy sample, a blood sample, a serum sample, a plasma sample, a saliva sample, a urinary sample, a lymph sample, or a bone marrow aspirate from the subject.
In preferred embodiments of the methods, the nucleic acids for use in the assay is genomic DNA.
Definition subject - recurrent or refractory form of a hyperproliferative disease
In preferred embodiments, the subject is diagnosed with or at risk of a recurrent or refractory form of a hyperproliferative disease, which subject has previously been treated with an irreversible BTK inhibitor.
At risk of a recurrent or refractory form of a hyperproliferative disease in the context of this invention is defined as the subject having a mutation in a gene that results in expression of a mutated BTK protein that is unable to bind a BTK inhibitor, preferably an irreversible BTK inhibitor.
In preferred embodiments, the subject is diagnosed with recurrent or refractory form of a hyperproliferative disease, preferably a B-cell haematological malignancy that is resistant, or non-responsive to therapy with an irreversible BTK inhibitor.
In preferred embodiments, the subject is identified as having a mutation in a gene that results in expression of a mutated BTK protein that is unable to bind a BTK inhibitor, preferably an irreversible BTK inhibitor, which provides a risk of a recurrent or refractory form of a hyperproliferative disease, preferably a B-cell haematological malignancy that is resistant, or non-responsive to therapy with an irreversible BTK inhibitor.
Definition relapse or refractory of disease
In preferred embodiments, the combination therapy is started when the B-cell haematological malignancy of the subject is relapsed or refractory, preferably wherein CLL/SLL is relapsed or refractory.
In preferred embodiments, the combination therapy is started when the B-cell haematological malignancy of the subject is relapsed or refractory and the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 , of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is C481 S.
Pharmaceutical composition
In a preferred embodiment, the pharmaceutical composition comprises a combination of an irreversible BTK inhibitor and a reversible BTK inhibitor, wherein the reversible BTK inhibitor is a compound according to any one of Formula (l-a) to (l-h), in a particular example a compound according to Formula A.
Pharmaceutical compositions in accordance with the present invention may comprise, as one of the active ingredients (‘API’), compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt, hydrate or solvate thereof.
As used herein, “a pharmaceutically acceptable salt” includes any salt that retains the activity of the active agent(s) and is acceptable for pharmaceutical use. A pharmaceutically acceptable salt also refers to any salt which may form in vivo as a result of administration of an acid, another salt, or a prodrug which is converted into an acid or salt. Preferably, the pharmaceutically acceptable salt is the HCI-salt of the compound of the invention. The pharmaceutically acceptable salt of the disclosed compounds may be prepared by methods of pharmacy well known to those skilled in the art.
Furthermore, the compositions can comprise compounds according to the invention in the form of a solvate, comprising a pharmaceutically acceptable solvent, such as water (‘hydrate’), ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
As used herein, the term “pharmaceutical composition” refers to a composition comprising a compound according to the invention or a salt or solvate thereof and, as the case may be, one or more additional, non-toxic ingredients, which composition is in a form suitable for administration to a (human) subject, through any route of administration, and which composition is physiologically tolerated upon such administration.
The compositions of the invention may thus comprise one or more additional ingredients. In a preferred embodiment, the composition comprises one or more carriers and/or excipients. As is known by those of average skill in the art, the appropriate choice of excipients is dependent on multiple factors, including the physicochemical properties of the API, the preferred pharmaceutical form, the preferred route of administration, the desired rate of release, etc. The compositions of the invention can be formulated for a variety of routes of administration, oral administration being particularly preferred. It is within the purview of those of average skill in the art to conceive and develop suitable formulations, relying on the common general knowledge as reflected in text books such as Remington’s Pharmaceutical Sciences (Meade Publishing Co., Easton, Pa., 20.sup.th Ed., 2000), the entire disclosure of which is herein incorporated by reference, and routine development efforts.
In accordance with the various aspects of the invention, the composition is preferably provided in a unit dosage form. The term ‘unit dosage form’ refers to a physically discrete unit suitable as a unitary dosage for human subjects, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with any suitable pharmaceutical carrier(s) and/or excipient(s). Exemplary, non-limiting unit dosage forms include a tablet (e.g., a chewable tablet), caplet, capsule (e.g., a hard capsule or a soft capsule), lozenge, film, strip, gelcap as well as any metered volume of a solution, suspension, syrup or
elixir orthe like, which may be contained, for instance in a vial, syringe, applicator device, sachet, spray, micropump etc. In accordance with particularly preferred embodiments of the invention, the unit dosage form, is a unit dosage form that is suitable for oral administration. Most preferably, it is a solid unit dosage form, such as a tablet.
Besides the compound according to the invention as such, pharmaceutically acceptable salts thereof may also be used. Pharmaceutically acceptable salts of compounds of the invention include the acid addition and base salts thereof, such as preferably the calcium, potassium or sodium salts. For a review on suitable salts, reference is made “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
Pharmaceutically acceptable salts of compounds according to the invention may be readily prepared by mixing together solutions of compounds according to the invention and the desired acid or base, as appropriate. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionisation in the salt may vary from completely ionised to almost non-ionised.
Medical Use
The compounds and the pharmaceutical compositions of the present invention are useful as inhibitors of tyrosine kinases, in particular of BTK. In particular, compounds of this invention are useful as inhibitors of tyrosine kinases that are important in hyper-proliferative diseases, especially in cancer, such as a B-cell hematological malignancy, and in the process of angiogenesis.
The reversible BTK inhibitor compounds according to the invention having any one of Formula (l-a) to (l-h) and pharmaceutical compositions comprising these, can be used to treat or prevent a variety of conditions, diseases or disorders mediated by Bruton’s Tyrosine kinase (BTK).
Such conditions, diseases or disorders include: cancers or tumors, including alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer including mast cell tumor and squamous cell carcinoma, breast and mammary cancer, ovarian cancer, prostate cancer, B cell hematological malignancy, lymphoma and leukemia (including but not limited to acute myelogenous leukemia, chronic myelogenous leukemia, mantle cell lymphoma, NHL B cell lymphomas (e.g. precursor B-ALL, marginal zone B-cell lymphoma, chronic lymphocytic leukemia, diffuse large B cell lymphoma, Burkitt lymphoma, mediastinal large B-cell lymphoma),
Hodgkin lymphoma, NK and T cell lymphomas, TEL-Syk and ITK-Syk fusion driven tumors, myelomas including multiple myeloma, myeloproliferative disorders kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer, brain cancer, melanoma including oral and metastatic melanoma, Kaposi’s sarcoma, proliferative diabetic retinopathy, and angiogenic- associated disorders including solid tumors, and pancreatic cancer.
In preferred embodiments, the hyperproliferative disease is a B-cell hematological malignancy.
In particular embodiments, the B-cell hematological malignancy is any one of chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), diffuse large B-cell lymphoma (DLBCL), activated B-cell diffuse large B-cell lymphoma (ABC-DLBCL), germinal center diffuse large B-cell lymphoma (GCB DLBCL), primary mediastinal B-cell lymphoma (PMBL), nonHodgkin lymphoma, Burkitt’s lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, precursor B-cell acute lymphoblastic leukemia, hairy cell leukemia, mantle cell lymphoma, B-cell prolymphocytic leukemia, lym- phoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis.
In preferred embodiments, the B-cell hematological malignancy is mantle cell lymphoma (MCL), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), diffuse large B- cell lymphoma (DLBCL), Waldenstrom macroglobulinemia (WM), follicular lymphoma (FL) and marginal zone lymphoma (MZL).
In preferred embodiments, the B-cell malignancy is Mantle cell lymphoma (MCL) or chronic lymphocytic leukemia (CLL).
The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks’s solution, Ringer’s solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
Pharmaceutical preparations for oral use can be obtained by combining the active compound with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from
pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The compounds can be formulated for parenteral administration by injection, e.g. bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g. in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly or by intramuscular injection). Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
An example of a pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The cosolvent system may be the VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-
solvent system (VPD:5W) consists of VPD diluted 1 :1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Many of the compounds of the invention may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms.
Synthesis of compounds
The compounds of the present invention can be prepared by methods well known in the art of organic chemistry. See, for example, J. March, ‘Advanced Organic Chemistry’ 4th Edition, John Wiley and Sons. During synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This is achieved by means of conventional protecting groups, such as those described in T.W. Greene and P.G.M. Wutts ‘Protective Groups in Organic Synthesis’ 3rd Edition, John Wiley and Sons, 1999. The protective groups are optionally removed at a convenient subsequent stage using methods well known in the art.
The products of the reactions are optionally isolated and purified, if desired, using conventional techniques, including but not limited to, filtration, distillation, crystallization, chromatography and the like. Such materials are optionally characterized using conventional means, including physical constants and spectral data. Compounds of any one of Formula l-a to l-h, wherein R1 to R4 and W, V and U have the previously defined meanings, can be prepared by the general synthetic route shown in any one of scheme I - XII, as described in co-pending PCT application PCT/EP2021/085641 and of the PCT application PCT/EP2022/085765.
Examples of synthesis of compounds
Benzyl (1 /?,5/?)-5-hydroxycyclohex-3-ene-1 -carboxylate
(a) (1 F?,4F?,5F?)-4-iodo-6-oxabicyclo[3.2.11octan-7-one
(R)-(+)-3-Cyclohexenecarboxylic acid (50.7 g, 402 mmol) was suspended in H2O (400 mL) under nitrogen. The mixture was cooled to 4 °C and sodium bicarbonate (101.3 g, 1 .21 mol) was added, followed by a solution of potassium iodide (333 g, 2.01 mol) and iodine (107 g, 422 mmol) in H2O (400 mL). The reaction was allowed to come to room temperature and stirred o/n and then extracted with dichloromethane (4x100 mL). The combined organic layers were washed with sat. NaHSOs-solution (2x50 mL). The organic layer was protected from light, dried over Na2SO4, filtered and concentrated (20 mbar) to afford (1 R,4/?,5/?)-4-iodo-6- oxabicyclo[3.2.1]octan-7-one (90.1 g, 89.0 %) as an off-white solid.
(b) (1 -6-oxabicyclo[3.2.11oct-3-en-7-one
(1 R,4/?,5/?)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (90.1 g, 358 mmol) was dissolved in dry THF (650 mL). Then, DBU (77 mL, 515 mmol) was added and the mixture was refluxed for 6 h. After cooling to room temperature, the suspension was filtered through Celite™, and concentrated in vacuo to -250 mL. This was used directly in the next step.
To the THF solution of (1 R,5/?)-6-oxabicyclo[3.2.1]oct-3-en-7-one (0.4 mol) in methanol (300 mL) was added 2M NaOH-solution (300 mL) and the mixture was stirred for 15 min at room temperature. The reaction was quenched by addition of 3M HCI-solution (300 mL) and the water layer was saturated by addition of sodium chloride. The mixture was extracted with ethyl acetate (3x100 mL). The combined organic phases were washed with brine, dried over sodium sulfate, filtered and the solvent removed in vacuo. The residue was dissolved in DMF (800 ml), cesium carbonate (129 g, 0.4 mol) and benzyl bromide (57 mL, 0.48 mol) were added subsequently. The mixture was stirred at room temperature for 30 min. The precipitate formed was filtered and the precipitate was washed with diethylether. The filtrate was washed with water, brine, dried over sodium sulfate and evaporated under reduced pressure. The residue was purified by
column chromatography (heptane/ethyl acetate = 95/5 to 45/55 v/v%) to give benzyl (1 R,5R)-5- hydroxycyclohex-3-ene-1-carboxylate (57.1 g, 61.5% over 3 steps) as a cream-coloured oil.
Ethyl (1 /?,5/?)-5-hydroxycyclohex-3-ene-1 -carboxylate -4-iodo-6-oxabicyclo[3.2.11octan-7-one
-3-Cyclohexenecarboxylic acid (50.7 g, 0.4 mol) was suspended in H2O (400 mL) under nitrogen. The reaction mixture was cooled to 4 °C and sodium bicarbonate (101 g, 1.2 mol) was added, followed by a solution of potassium iodide (333 g, 2 mol) and iodine (107 g, 0.42 mol) in H2O (400 mL). The reaction was allowed to come to room temperature and stirred o/n and then extracted with DCM (4x150 mL). The combined organic layers were washed with a solution of Na2S20s (120 g) in H2O (600 mL). The aqueous layer was extracted with DCM (2x150 mL). The combined organic layers were protected from light, dried over Na2SO4, filtered and concentrated (20 mbar) to afford (1 R,4/?,5/?)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (95.22 g, 94.5 %) as an off-white solid.
(b) (1 f?,5f?)-6-oxabicyclo[3.2.11oct-3-en-7-one
(1 R,4/?,5/?)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (95.22 g, 377.9 mmol) was dissolved in dry THF (700 mL). Then, DBU (86.3 g, 566.9 mmol) was added and the mixture was refluxed o/n. The reaction mixture was cooled to room temperature, diluted with diethylether (500 mL) and extracted with aq. HCI (1 L, 1 M) and brine (250 mL). The aqueous layers were extracted with diethylether (2 x 480 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated (350 mbar to afford (1 R,5/?)-6-oxabicyclo[3.2.1]oct-3-en-7-one quantitatively as a yellowish oil which was used directly in the next step.
To a stirred solution of (1 R,5/?)-6-oxabicyclo[3.2.1]oct-3-en-7-one (377.9 mmol, theor.) in ethanol (750 mL) was added potassium carbonate (10.45 g, 75.6 mmol) at room temperature and the mixture stirred o/n. The reaction mixture was filtered through a Celite pad. Removal of ethanol under reduced pressure afforded the crude product that was purified by column chromatography plug filtration (eluent 40% EtOAc/heptane) to afford the title compound (41.38 g, 60.8% over 3 steps and column) as a yellow liquid.
Intermediate 3
Methyl (1 /?,5/?)-5-hydroxycyclohex-3-ene-1 -carboxylate
(R)-(+)-3-Cyclohexenecarboxylic acid (20.2 g, 160 mmol) was suspended in H2O (430 mL) under nitrogen. The reaction mixture was cooled to 0 °C and sodium bicarbonate (40.3 g, 480.3 mmol) was added, followed by a solution of potassium iodide (159.5 g, 961 mmol) and iodine (39.6 g, 168 mmol) in H2O (360 mL). The reaction was allowed to come to room temperature and stirred o/n and then extracted with DCM (3x150 mL). The combined organic layers were washed with a solution of Na2S20s (120 g) in H2O (600 mL). The aqueous layer was extracted with DCM (2x150 mL). The combined organic layers were protected from light, dried over Na2SO4, filtered and concentrated (20 mbar) to afford (1 R,4/?,5/?)-4-iodo-6- oxabicyclo[3.2.1]octan-7-one (37.88 g, 93.9 %) as an off-white solid.
(b) (1 f?,5f?)-6-oxabicyclo[3.2.11oct-3-en-7-one
(1 R,4/?,5/?)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (37.88 g, 150.3 mmol) was dissolved in dry THF (750 mL). Then, DBU (34.3 g, 225.2 mmol) was added and the mixture was refluxed o/n. The reaction mixture was cooled to room temperature, diluted with diethylether (480 mL) and extracted with aq. HCI (1 L, 0.5 M) and brine (1 L). The aqueous layers were extracted with diethylether (2 x 480 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated (350 mbar to afford (1 R,5/?)-6-oxabicyclo[3.2.1]oct-3-en-7-one quantitatively as a yellowish oil which was used directly in the next step. -5-hydroxycyclohex-3-ene-1 -carboxylate (Intermediate 3)
icarbonate (37.88 g, 0.451 mol) was added to a solution (1 R,5/?)-6- oxabicyclo[3.2.1]oct-3-en-7-one (150.3 mmol theor.) in anhydrous MeOH (300 mL). After stirring for 1 week at room temperature the solvent was removed in vacuo (40 °C/300 mbar). The residue was diluted with water (500 mL) and extracted with dichloromethane (3x250 mL). The combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (21 .2 g, 90.3%) as a slightly coloured liquid.
Intermediate 4 (Route A)
(1 /?,3/?)-3-(Benzyloxycarbonylamino)cvclohexanecarboxylic acid
(a) Methyl (1 /?,5S)-5-[bis(te/Y-butoxycarbonyl)amino1cyclohex-3-ene-1 -carboxylate
To an ice-cold (4 °C) solution of di-te/Y-butyl iminodicarboxylate (4.6 g, 21 .2 mmol), methyl (1 R,5/?)-5-hydroxycyclohex-3-ene-1 -carboxylate (Intermediate 3) (3.31 g, 21.2 mmol) and triphenylphosphine (6.67 g, 25.4 mmol) in 2-MeTHF (180 mL) was added dropwise a solution of diisopropyl azodicarboxylate (6.26 mL, 31 .8 mmol) in 2-MeTHF (30 mL). The mixture was stirred for 30 min at 4 °C and then allowed to warm to room temperature and stirred for 3 h. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography using SiO2 and heptane/ethyl acetate = 3/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 5.68 g of methyl (1 R,5S)-5- [bis(te/Y-butoxycarbonyl)amino]cyclohex-3-ene-1 -carboxylate (yield 75.4 %).
(b) Methyl (1 F?,3F?)-3-[bis(te/Y-butoxycarbonyl)amino1cvclohexanecarboxylate
To a solution of methyl (1 /?,5S)-5-[bis(te/Y-butoxycarbonyl)amino]cyclohex-3-ene-1- carboxylate (2.48 g, 6.98 mmol) in ethanol (140 mL) was added 248 mg of 10% Pd/C. Catalytic hydrogenation was performed at room temperature for 16 h. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to give 2.6 g of the title compound (Yield: quantitative).
(c) Methyl (1 F?,3F?)-3-aminocvclohexanecarboxylate hydrochloride
To methyl (1 /?,3/?)-3-[bis(te/Y-butoxycarbonyl)amino]cyclohexanecarboxylate (2.6 g, 7.2 mmol) was added 4M HCI/dioxane solution (18 mL) and the mixture was stirred at room temperature o/n. The mixture was concentrated in vacuo to give 1 .06 g the title compound (yield: 76%).
(d) Methyl (1 F?,3F?)-3-(benzyloxycarbonylamino)cvclohexanecarboxylate
Methyl (1 R,3/?)-3-aminocyclohexanecarboxylate hydrochloride (1.06 g, 5.47 mmol) was suspended in 10 mL water. Sodium bicarbonate (1.38 g, 16.4 mmol) in 10 mL water was added followed by a drop-wise addition of a solution A/-(benzyloxycarbonyloxy)succinimide (1 .50 g, 6.01 mmol) in dioxane (30 mL). The reaction mixture was stirred at room temperature o/n. The mixture was diluted with ethyl acetate (50 mL) and water (50 mL) and the bi-phasic system was stirred
30 minutes at room temperature. The layers were separated and the water layer was extracted with ethyl acetate (2x20 mL). The combined organic layers were washed with water (50 mL), 0.5N aq. HCI-solution (50 mL), water (50 mL), 5% aq. NaHCOs-solution (50 mL), water (50 mL) and brine (25 mL), dried (Na2SO4), filtered and concentrated in vacuo to give 1.78 g of the title compound (yield: quantitative, crude).
(e) (1 f?,3f?)-3-(Benzyloxycarbonylamino)cvclohexanecarboxylic acid (Intermediate 4)
The crude product methyl (1 R,3/?)-3-(benzyloxycarbonylamino)cyclohexanecarboxy- late (1.45 g, 4.98 mmol) was dissolved in THF/dioxane/water = 4/1/1 v/v% (74 mL) and subsequently lithium hydroxide (358 mg, 14.9 mmol) was added. The mixture was stirred at room temperature o/n. Ethyl acetate (50 mL) and water (were added) and the pH of the mixture was adjusted to pH < 3 by addition of 2M HCI-solution. The organic phase was separated, washed with water, brine, dried over sodium sulfate , filtered and concentrated under reduced pressure to give 800 mg of (1 R,3/?)-3-(benzyloxycarbonylamino)cyclohexanecarboxylic acid (Intermediate 4) (yield:57.9%).
-3-(Benzyloxycarbonylamino)cvclohexanecarboxylic acid
(a) tert-Butyl A/-(4-nitrophenyl)sulfonylcarbamate
Triethylamine (10.4 mL, 74.62 mmol), 4-dimethylaminopyridine (605 mg, 4.95 mmol) di- fe/Y-butyl dicarbonate (13.5 g, 61.86 mmol) were added sequentially to a solution of 4- nitrobenzene sulfonamide (10 g, 49.46 mmol) in dichloromethane (100 mL). The reaction mixture was stirred for 30 minutes at room temperature. To the reaction mixture was added hydrochloric acid (1 N aqueous solution) until it becomes acidic. The organic layer was separated and washed
with saturated sodium chloride aqueous solution, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residue was dissolved in dichloromethane and purified by column chormatography over silica (heptane to ethyl acetate = 10/0 to 0/10) to give 13.09 g of the title compound (yield: 87.5%).
(b) Methyl (1 f?,5S)-5-[te/Y-butoxycarbonyl-(4-nitrophenyl)sulfonyl-amino1-cvclohex-3-ene-1 - carboxylate
To a cold (-20 °C) solution of methyl (1 R,5/?)-5-hydroxycyclohex-3-ene-1 -carboxylate (15 g, 96.0 mmol), te/Y-butyl A/-(4-nitrophenyl)sulfonylcarbamate (29.0 g, 96.0 mmol) and triphenylphosphine (27.7 g, 105.6 mmol) in THF (300 mL) was added dropwise a solution of diisopropyl azodicarboxylate (20.8 mL, 105.6 mmol) in THF (100 mL). The reaction mixture was concentrated under reduced pressure to give a residue which was purified by column chromatography (hexane/ethyl acetate = 85/15 v/v%) to give 36 g of the title compound, (yield: 85.1 %).
(c) Methyl (1 F?,5S)-5-(tert-butoxycarbonylamino)cyclohex-3-ene-1 -carboxylate
To a stirred solution of methyl (1 R,5S)-5-[te/Y-butoxycarbonyl-(4-nitrophenyl)sulfonyl- amino]-cyclohex-3-ene-1 -carboxylate (35.15 g, 79.8 mmol) in acetone (300 mL) was added DBU (23.85 mL, 159.6 mmol) and 2-mercaptoethanol (11.23 mL, 159.6 mmol). The reaction mixture was stirred for 3 h at room temperature. Acetone was removed under reduced pressure and the resulting residue was purified by column chromatography (hexane/ethyl acetate = 90/1 O to 88/12 v/v%) to give 14.1 g of the title compound (Yield: 69.2%) as a crystalline white solid.
(d) Methyl (1 F?,3F?)-3-(te/Y-butoxycarbonylamino)cvclohexanecarboxylate
To a solution of methyl (1 R,5S)-5-(tert-butoxycarbonylamino)cyclohex-3-ene-1- carboxylate (14.9 g, 58.36 mmol) in methanol (300 mL) was added 1 .5 g of 10% Pd/C. Catalytic hydrogenation was performed for 3 h at room temperature. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to afford the title compound in quantitative crude yield.
(e) Methyl (1 F?,3F?)-3-aminocvclohexanecarboxylate hydrochloride
To a cooled (4 °C) solution of methyl (1 /?,3R)-3-(te/Y-butoxycarbonylamino)cyclo- hexanecarboxylate (15.2 g, 58.36 mmol) in methanol (300 mL) was added drop-wise acetyl chloride (42 mL, 583.6 mmol). The reaction mixture was stirred for 1 h. The mixture was concentrated under reduced pressure and dried in vacuo to give the title compound in quantitative crude yield.
(f) Methyl (1 F?,3F?)-3-(benzyloxycarbonylamino)cvclohexanecarboxylate
To a cooled (4 °C) solution of methyl (1 R,3/?)-3-aminocyclohexanecarboxylate hydrochloride (58.36 mmol) in dioxane/water = 1/1 v/v% (200 mL) was added portion-wise sodium bicarbonate (14.7 g, 175 mmol). To the resulting suspension was added drop-wise a solution of A/-(benzyloxycarbonyloxy)succinimide (14.8 g, 59.02 mmol) in dioxane (150 mL) and the resulting mixture was stirred at room temperature o/w. LC-MS showed some starting material present. Additionally, 1 .5 g of Z-ONSu was added as a solution in dioxane. The mixture was stirred at room temperature o/n. Ethyl acetate was added and the resulting mixture was washed
with 0.5M HCI solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and concentrated under reduced pressure and dried in vacuo to give the title compound in quantitative crude yield.
(g) (1 R,3R)-3-(benzyloxycarbonylamino)cvclohexanecarboxylic acid (Intermediate 4)
To a solution of methyl (1 R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxylate (58.36 mmol) in THF/water = 4/1 v/v% (375 mL) was added lithium hydroxide (4.21 g, 175 mmol) and the reaction mixture was stirred at room temperature o/n. Ethyl acetate (300 mL) and water (300 mL) were added to the mixture and the aqueous phase was separated. The ethyl acetate layer washed extracted with water (100 mL). The combined aqueous phases were washed with dichloromethane (100 mL) and acidified (pH < 2) by addition of 2M HCI-solution (90 mL). The water layer was extracted with ethyl acetate (3x250 mL). The combined ethyl acetate layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure and dried in vacuo to give 15.72 g of the title compound (Yield: 96.7% over 4 steps).
(1 -3-(te/Y-butoxycarbonylaminocvclohexanecarboxylic acid
(a) Ethyl (1 R,5S)-5-(1 ,3-dioxoisoindolin-2-yl)cyclohex-3-ene-1 -carboxylate
To an ice-cold (0 °C) solution of ethyl (1 R,5R)-5-hydroxycyclohex-3-ene-1 -carboxylate (Intermediate 2, 15.0 g, 88.13 mmol), phthalimide (14.26 g, 96.94 mmol) and triphenylphosphine (34.67 g, 132.2 mmol) in toluene (264 mL) was added dropwise diisopropyl azodicarboxylate (26.02 mL, 132.2 mmol) in 10 min. The reaction mixture was stirred at 0 °C for 30 min and then allowed to come to room temperature and stirred for 3 h. The mixture was evaporated under reduced pressure to give a yellow oil. Heptane/ethyl acetate = 7/3 v/v% (500 mL) was added and the mixture was heated to 70 °C. After cooling, the mixture was stirred for 72 h at room temperature. The solids were filtered, washed with heptane/ethyl acetate = 9/1 v/v% (2x50 mL) and the filtrate was evaporated under reduced pressure. The resulting residue was purified by
column chromatography (heptane/ethyl acetate = 9/1 to 6/4 v/v%) to give 21 .96 g of the title compound (Yield: 83.0%) as an off-white solid.
(b) Ethyl (1 F?,3F?)-3-(1 ,3-dioxoisoindolin-2-yl)cvclohexanecarboxylate
To a solution of ethyl (1 R,5S)-5-(1 ,3-dioxoisoindolin-2-yl)cyclohex-3-ene-1 -carboxylate (27.73 g, 97.19 mmol) in methanol (975 mL) was added 2.7 g of 10% Pd/C. Catalytic hydrogenation was performed for 3 h at room temperature. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to afford 27.52 g the title compound (Yield: 94%).
(c) Ethyl (1 f?,3f?)-3-aminocvclohexanecarboxylate
To a solution of ethyl (1 R,3/?)-3-(1 ,3-dioxoisoindolin-2-yl)cyclohexanecarboxylate (26.5 g, 87.95 mmol) in ethanol (440 mL) was added drop-wise hydrazine hydrate (64% in water, 4.68 mL, 96.74 mmol). The reaction mixture was stirred for 30 min. at room temperature and then refluxed for 3 h. Additional hydrazine hydrate (425 pL) was added and stirring under reflux was continued for 2 h. The mixture was concentrated under reduced pressure and dried in vacuo to give the title compound in quantitative crude yield.
(d) Ethyl (1 f?,3f?)-3-(te/Y-butoxycarbonylamino)cvclohexanecarboxylate
To a cold (0 °C) stirred suspension of ethyl (1 R,3/?)-3-aminocyclohexanecarboxylate (91.27mmol, theoretical) in dichloromethane (456ml) was added portion-wise di-tert-butyl dicarbonate (21 .91 g, 100.4 mmol). The reaction mixture was stirred for 15 min at 0 °C, then allowed to come to room temperature. The mixture was washed with cold 0.5N NaOH-solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (heptane/ethyl acetate = 8/2 to 6/4 v/v%) to give 20.5 g of the title compound (Yield: 83.0% over two steps) as an off-white solid.
(e) (1 f?,3f?)-3-(te/Y-butoxycarbonylamino)cvclohexanecarboxylic acid
To a solution of ethyl (1 /?,3R)-3-(te/Y-butoxycarbonylamino)cyclohexanecarboxylate (20.5 g, 75.56 mmol) in THF (300 mL) was added a solution of lithium hydroxide (1.8 g, 75.56 mmol) in water (150 mL) and the reaction mixture was stirred at room temperature o/n. Additional lithium hydroxide (0.9 g) was added and stirring was continued for 24 h at room temperature. The mixture was acidified (pH < 2) by addition of 1 M HCI-solution (131 mL). The water layer was separated and extracted with dichloromethane (2x100 mL). The combined organic layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure and dried in vacuo to give 16.81 g (91 %) of (1 /?,3R)-3-(te/Y-butoxycarbonylamino)cyclo- hexanecarboxylic acid (Intermediate 5).
(6-Bromo-3-chloro-pyrazin-2-yl)methanamine hydrochloride
(a) 3-Amino-6-bromo-pyrazine-2-carbonitrile
To a solution of commercially available 2-amino-3,5-dibromo pyrazine (25.12 g, 99.3 mmol) in DMF (160 mL) was added sodium cyanide (4.97 g, 101.3 mmol) and copper(l)cyanide (9.07 g, 101 .3 mmol) at room temperature. The mixture was stirred at 95 °C for 32 h. After cooling the mixture was and poured in a thoroughly stirred mixture of 5% sodium bicabonate solution (1 L) and ethyl acetate (500 mL) to give a black suspension. The mixture was filtered over Decalite®, flushed with warm ethyl acetate (2 x 150 mL). The organic layer from the biphasic filtrate was separated. The waterlayer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to give 15.6 g of the crude title compound (yield: 78.9%)
(b) 6-Bromo-3-chloro-pyrazine-2-carbonitrile
To a solution of 3-amino-6-bromo-pyrazine-2-carbonitrile (15.6 g, 78.4 mmol) in acetonitrile (350 mL) was added copper(ll)chloride (12.65 g, 94.1 mmol) and the mixture was heated to 60 °C and stirred for 15 min. A solution of tert-butyl nitrite (1 1.75 mL, 98 mmol) in acetonitrile (50 mL) was added dropwise at 60 °C to the reaction mixture. The mixture was stirred at 60 °C o/n. After cooling, the mixture was filtered over Decalite®, washed with acetonitrile. The filtrate was added dropwise to a 1 N HCI-solution (250 mL) and ethyl acetate (250 mL) and stirred for 10 min. The layers were separated and the waterlayer was extracted with ethyl acetate (2x 100 mL). The combined organic layers were washed with 1 N-HCl-solution, water and brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (plug filtration) using SiO2 and heptane/ethyl acetate = 95/5 to 9/1 to 8/2 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 10.4 g of the title compound (yield 60.7%).
(c) (6-Bromo-3-chloro-pyrazin-2-yl)methanamine hydrochloride
To a solution of 6-bromo-3-chloro-pyrazine-2-carbonitrile (10.4 g, 47.62 mmol) in acetic acid (160 mL) was added under nitrogen of Raney-Nickel (50% slurry in water, 1 .96 mL, -16.7 mmol). The resulting mixture was shaken in a Parr-vessel under 4 bar hydrogen at room temperature o/n. Raney-Nickel was removed by filtration over Decalite® and the filtrate was concentrated under reduced pressure and co-evaporated with toluene. The remaining green solid (12.6 g) was suspended in ethyl acetate at 50 °C and filtered. The filtrate was cooled on an ice-bath, 2N HCIgas in diethyl ether (48 mL) was added dropwise and the resulting suspension was stirred at room temperature o/n. The solids formed were collected by filtration, washed with diethyl ether and dried under reduced pressure at 40 °C to give 6.21 g (50%) of (6-bromo-3- chloro-pyrazin-2-yl)methanamine hydrochloride (Intermediate 6).
Ethyl (E)-8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)oct-7-enoate (Intermediate 7)
(a) Ethyl oct-7-ynoate
To a solution of oct-7-ynoic acid (7.92g, 56.53 mmol) in ethanol (70 ml) was added conc.- H2SO4 (0.2 mL). The reaction mixture was stirred for 5 h. at 70 °C. After cooling of the mixture, MTBE (150 mL) was added and the organic phase was washed with sat. aq. NaHCOs-solution, water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 8.84 g of ethyl oct-7-ynoate (yield: 93%).
(b) Ethyl (E)-8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)oct-7-enoate (Intermediate 7)
To a cold (5 °C) oven-dried round bottom flask equipped with a magnetic stirring bar were added ethyl oct-7-ynoate (9.11 g, 54.16 mmol), Schwartz’s reagent (1.39 g, 5.41 mmol), EtaN (753 pL, 5.41 mmol) and pinacolborane (8.24 mL, 56.86 mmol), under an inert nitrogen atmosphere. The bottom flask was then sealed and the mixture was stirred at 60 °C for 5 hours. The reaction was allowed to cool to room temperature, diluted with dichloromethane, passed through a pad of silica gel and concentrated under reduced pressure at room temperature. The crude mixture was purified by column chromatography using SiO2 and hexane/ethyl acetate = 99/1 to 9/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 6.42 g of the title compound (yield: 40%).
A/-(4-Cyano-2-pyridyl)-4-(4,4,5,5-tetramethyl-1 , 3, 2-dioxaborolan-2-yl) benzamide (I ntermed iate 8)
(a) 4-(4,4,5,5-Tetramethyl-1 , 3, 2-dioxaborolan-2-yl) benzoyl chloride
To a cold (0 °C) solution of 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)benzoic acid (24.8 g, 100 mmol) in dichloromethane (300 mL) was added a catalytic amount of DMF. A solution of oxalyl chloride (12.9 mL, 150 mmol) was added dropwise. After stirring for 60 min at
0 °C, the reaction mixture was allowed to warm to room temperature and the mixture was stirred o/n. The reaction mixture was concentrated to give 26.33 g of crude 4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)benzoyl chloride (yield: 99%).
(b) A/-(4-Cyano-2-pyridyl)-4-(4,4,5,5-tetramethyl-1 , 3, 2-dioxaborolan-2-yl) benzamide (Intermediate 8)
To a cold (0 °C) solution of 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)benzoyl chloride (26.0 g, 100 mmol) in acetonitrile (300 mL) was subsequently added 2-aminopyridine- 4-carbonitrile (14.29 g, 120 mmol) and 4-DMAP (14.66 g, 120 mmol) The mixture was stirred under nitrogen atmosphere at 0°C and allowed to warm to room temperature overnight. The reaction mixture was concentrated in vacuo. The crude oily solids were then dissolved in dichloromethane (300 mL) and washed with 5% citric acid (3x, 300 mL), 5% NaHCOs (2x300 mL) and brine (200 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The residue triturated in acetonitrile/water = 1/1 v/v% (300 mL). The solids were filtrated and dried under reduced pressure to give 20.5 g of 4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-A/-[4-(trifluoromethyl)-2-pyridyl]benzamide (Intermediate 8) (yield: 69%) as off-white solids.
(a) te/Y-Butyl A/-[(1 /?,3/?)-3-[(6-bromo-3-chloro-pyrazin-2-yl)methylcarbamoyl1cvclohexyl1- carbamate
To a cold (4 °C) suspension of (1 /?,3R)-3-(te/Y-butoxycarbonylamino)cyclohexane- carboxylic acid (Intermediate 5, 10.36 g, 42.62 mmol) and (6-bromo-3-chloro-pyrazin-2- yl)methanamine hydrochloride (intermediate 6, 10.51 g, 40.59 mmol) in dichloromethane (406 mL) was added subsequently, DiPEA (21 .21 mL, 121 .77 mmol) and HATU (16.2 g, 42.62 mmol). The resulting mixture was stirred at room temperature o/n. The reaction mixture was filtered over a Buechner filter. The filtrate was washed with 5% aq. NaHCOs-solution (100 mL), 5% aq. citric acid solution (3x100 mL), brine (100 mL), dried over sodium sulfate filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO2 and heptane/ethyl acetate = 8/2 to 3/7 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 17.4 g of the title compound (Yield: 94%).
(b) Ethyl (E)-8-[6-[[[(1 f?,3f?)-3-(te/Y-butoxycarbonylamino)cvclohexanecarbonyl1amino1methyl1- 5-chloro-pyrazin-2-yl1oct-7-enoate
A mixture of te/Y-butyl A/-[(1 R,3R)-3-[(6-bromo-3-chloro-pyrazin-2-yl)methylcarbamoyl]- cyclohexyl]carbamate (17.4 g, 38.86 mmol), ethyl (E)-8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)oct-7-enoate (Intermediate 7, 1 1.51 g, 38.86 mmol) and potassium carbonate (16.11 g, 116.6 mmol) in dioxane/water = 4/1 v/v% (200 mL) was degassed with nitrogen for 5 min at 30 °C. Pd(dppf)Cl2.CH2Cl2 (1.58 g, 1 .94 mmol) was added and the mixture was again degassed with nitrogen for 5 min at 30 °C. The reaction mixture was stirred at 80 °C for 4 h. After cooling ethyl acetate was added and the mixture was stirred for 15 min. The mixture was filtered over Decalite™ and the filtrate was washed with water and brine. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The crude product was purified by column chromatography using SiO2 and heptane/ethyl acetate = 7/3 to 3/7 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 20.9 g of the title compound (Yield: quantitative).
(c) Ethyl (E)-8-[3-[(1 -3-(te/Y-butoxycarbonylamino)cvclohexyl1-8-chloro-imidazo[1 ,5-
al py razi n -5-v II o ct-7-e n o ate
To a cold (-10 °C) solution of ethyl (E)-8-[6-[[[(1 R,3R)-3-(te/Y-butoxycarbonylamino)- cyclohexanecarbonyl]amino]methyl]-5-chloro-pyrazin-2-yl]oct-7-enoate (20.9 g, 38.9 mmol) in acetonitrile/DMF = 9/1 v/v% (117 mL) was added subsequently pyridine (12.58 mL, 155.7 mmol) and phosphorus oxychloride (7.25 mL, 77.84 mmol) and the mixture was stirred for 30 min. at 5 °C. The mixture was added carefully to 25% ammonia (200 mL) and crushed ice (400 mL) keeping the temperature below 0 °C. Ethyl acetate (200 mL) was added and the resulting mixture was stirred for 30 min. The water layer was separated and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated in vacuo to give 19.4 g of the title compound (yield: 96%).
(d) Ethyl (E)-8-[1-bromo-3-[(1 /?,3/?)-3-(te/Y-butoxycarbonylamino)cvclohexyl1-8-chloro-imidazo- [1 ,5-a1pyrazin-5-yl1oct-7-enoate
/V-Bromosuccinimide (7.31 g, 41.1 mmol) was added to a cold (0 °C) solution of ethyl (E)-8-[3-[(1 /?,3/?)-3-(te/Y-butoxycarbonylamino)cyclohexyl]-8-chloro-imidazo[1 ,5-a]pyrazin-5- yl]oct-7-enoate (19.4 g, 37.37 mmol) in acetonitrile (149 mL). The reaction mixture was stirred for 3 h. allowing the temperature to come to room temperature. The mixture was quenched with 10% aq. Na2S2O4-solution/5% aq. NaHCOs-solution/brine and ethyl acetate. The phases were separated and the water layer was extracted with ethyl acetate. The combined organic phases were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO2 and heptane/ethyl acetate = 9/1 to 6/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 17.07 g of the title compound (Yield: 74%).
(e) Ethyl (E)-8-[1-bromo-3-[(1 -3-(tert-butoxycarbonylamino)cvclohexyl1-8-[(2,4-
dimethoxyphenyl)methylamino1imidazo[1 ,5-a1pyrazin-5-yl1oct-7-enoate
To a suspension of ethyl (E)-8-[1-bromo-3-[(1 /?,3R)-3-(te/Y-butoxycarbonylamino)- cyclohexyl]-8-chloro-imidazo-[1 ,5-a]pyrazin-5-yl]oct-7-enoate (17.07 g, 28.55 mmol) in 1 -butanol (114 mL) was added 2,4-dimethoxybenzylamine (12.86 mL, 85.65 mmol) and the mixture was stirred at 110 °C for 2 h. The mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate/water 5/2 v/v% (700 mL). The organic layers was separated and washed with 5% NaHCOs-solution (100 mL) and brine (2x50 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO2 and heptane/ethyl acetate = 85/15 to 6/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 18.9 g of the title compound (Yield: 91 %).
(f) Ethyl (E)-8-[3-[(1 /?,3/?)-3-aminocvclohexyl1-1-bromo-8-[(2,4-dimethoxyphenyl)methylamino1- imidazo[1 ,5-a1pyrazin-5-yl1oct-7-enoate
Ethyl (E)-8-[1 -bromo-3-[(1 /?,3/?)-3-(tert-butoxycarbonylamino)cyclohexyl]-8-[(2,4-dime- thoxyphenyl)methylamino]imidazo[1 ,5-a]pyrazin-5-yl]oct-7-enoate (18.85 g, 25.86 mmol) was dissolved in dichloromethane (129 mL). Trifluoroacetic acid (19.2 mL) was added and the mixture was stirred at room temperature for 2 h. The mixture was poured into dichloromethane/water = 1/4 v/v% (750 mL), the water layer was separated and extracted with dichloromethane (100 mL). The combined organic layers were washed with sat. aq. NaHCOs- solution (100 mL), brine (50 mL), dried over sodium sulfate, filtered and concentrated in vacuo to give 16.68 g of the title compound (Yield: 102%).
(g) Lithium (E)-8-[3-[(1 f?,3f?)-3-aminocvclohexyl1-1 -bromo-8-[(2,4-dimethoxyphenyl)methyl- amino1imidazo[1 ,5-a1pyrazin-5-yl1oct-7-enoate
To a solution ethyl (E)-8-[3-[(1 /?,3/?)-3-aminocyclohexyl]-1-bromo-8-[(2,4-dimethoxy- phenyl)methylamino]-imidazo[1 ,5-a]pyrazin-5-yl]oct-7-enoate (16.68 g, 25.86 mmol) in THF/water=4/1 v/v% (129 mL) was lithium hydroxide (648 mg, 27.15 mmol) and the mixture was
stirred at room temperature for 72 h. Water (300 mL) was added to the mixture and after evaporation of THF, the mixture was lyophilized to give 15.9 g of the title compound (yield: quantitative crude).
(h) Macrocyclic Intermediate 9
Lithium (E)-8-[3-[(1 R,3/?)-3-aminocyclohexyl]-1 -bromo-8-[(2,4-dimethoxyphenyl)- methylamino]imidazo[1 ,5-a]pyrazin-5-yl]oct-7-enoate (9.14 g, 15.07 mmol) was suspended dissolved in DMF (520 mL) and A/-ethylmorpholine (3.83 mL, 30.14 mmol) was added. This solution was added slowly to a heavily stirred suspension of HATU (17.18 g, 45.21 mmol) and A/-ethylmorpholine (5.74 mL, 45.21 mmol) in ethyl acetate (1 .0 L) over a 11 h period to obtain a clear solution. The mixture was poured into stirred cold water (2.0 L) and stirring was continued for 15 min. The water layer was separated and extracted with ethyl acetate/heptane = 95/5 v/v% (3x300 mL). The combined organic layer was washed with washed with 2.5% aq. citric acid solution (300 mL), sat. aq. NaHCOs-solution (300 mL), water (300 mL) and brine (100 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography using SiO2 and heptane/ethyl acetate/methanol = 50/50/0 to 0/92.5/7.5 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 7.39 g of the DMB-protected Macrocyclic Intermediate 9 (Yield: 84%).
(i) Example A: Compound A
DMB-protected Macrocyclic Intermediate 9 (9.83 g, 16.87 mmol) was dissolved in TFA (67 mL) and the mixture was stirred at 60 °C for 4 h. The reaction mixture was added slowly to a mixture of dichloromethane/water = 1/1 v/v% (1.0 L). The bi-phasic mixture was stirred for 10 min. The layers were separated and the water layer was extracted with dichloromethane (200 mL). To the combined organic layer was added slowly under stirring 12.5% NH4OH (300 mL). The layers were separated and the organic layer was washed with 12.5% NH4OH (150 mL), water (250 mL) and brine (100 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was triturated with hot (90 °C) acetonitrile (100 mL), filtered and dried to give 5.9 g (yield: 80%) of the deprotected Macrocyclic Intermediate 9 which was used directly in the next step.
Deprotected Macrocyclic Intermediate 9 (2.25 g, 5.2 mmol) was dissolved in dioxane/water = 9/1 v/v% (100 mL) and potassium carbonate (2.15 g, 15.6 mmol) was added. The solution was purged with nitrogen for 5 min and A/-(4-cyano-2-pyridyl)-4-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)benzamide (Intermediate 8) (2.18 g, 6.24 mmol) and Pd(dppf)Cl2.CH2Cl2 (424 mg, 0.52 mmol) were added. The reaction mixture was stirred for 4 h at 90 °C. The reaction mixture was diluted with dichloromethane/methanol = 9/1 v/v% (400 mL) after cooling to room temperature and filtered over Decalite™. The filtrate was washed with water, and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO2 and dichloromethane/methanol = 98/2 to 94/6 v/v%. All fractions containing the title compound were
collected and concentrated in vacuo to give 1 .6 g of the title compound (Yield: 53%). Data: LCMS (B) Rt : 6.99 min; m/z 575.4 [M+H]+.
Remark: Compound A as described herein is the same compound as compound having subformula 184 of co-pending PCT application PCT/EP2022/085765. Said compound having subformula 184 is also incorporated by reference.
Examples
Cell culture
TMD8 diffuse large B-cell lymphoma cells were purchased from Tokyo Medical and Dental University. TMD8 cells expressing mutant BTK were created at Synthego Corporation. TMD8 cell lines expressing BTK C481 S, BTK T474I, BTK T474I/C481S, BTK V416L and BTK L528W were generated via CRISPR/Cas9. BTK T474I/C481 S expressing cells were generated by re- editing BTK C481 S cells. TMD8 mutant cell pools were enriched to obtain a 100% mutant genotype. The mutation status of all cell lines was confirmed via sequencing. Cells were cultured in MEM-alpha cell culture medium (cat. no. 22571038, Life Technologies), supplemented with 10% (v/v) heat-inactivated fetal bovine calf serum (Avantor, cat. no. 97068085) and 1 % penicillin/streptomycin.
Proliferation assays
Inhibition of proliferation in response to compound was measured using the ATPIite 1 Step™ assay (cat. no. 6016736, Perkin Elmer). The assay utilizes ATP as a marker for metabolically active cells. A decline in ATP concentration indicates that compounds exert anti-proliferative effects on the cells under investigation, compared to vehicle treated cells. Measurement of ATP is based on the production of light caused by the reaction of ATP with added luciferase and D- luciferin. Luminescence was recorded using an Envision multilabel reader (Perkin Elmer). The ATP signal of the cells at the start of incubation (t=0) was recorded separately to distinguish between cell growth and cell death. Maximum growth was determined via vehicle treated cells, which indicates 100% viability (vehicle control). Percentage viability was used as the main y-axis signal. IC50S were fitted by non-linear regression using IDBS XLfit ™ 5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC50. In addition to the IC50, efficacy, GI50 and LD50 were also determined.
Proliferation assays on BTK single mutant cell lines
WT-BTK, BTK C481 S, BTK T474I, BTK T474I/C481 S, BTK V416L and BTK L528W TMD8 cell lines were seeded at 800 cells per well in a white 384-well culture plate (cat. no. 781080, Greiner Bio-One) and allowed to rest for at least 4 hours at 37 °C, 95 % humidity, and 5 % CO2.
Compound A, pirtobrutinib, ibrutinib, acalabrutinib and nemtabrutinb were diluted in 3.16-fold dilution steps in 100% DMSO and further diluted in 20 mM Hepes, generating a dose response range from 316 nM to 0.0316 nM in the assay. DMSO was used as a vehicle control. Luminescence was recorded at 96 hours.
Surface plasma resonance
Binding kinetics measurements on wt-BTK, BTK C481 S, BTK T474I (Surface Plasmon Resonance) Streptavidin-coated chips (Cat. No. BR100531), disposables and maintenance kits for Biacore were purchased from Cytiva (Eindhoven, The Netherlands). Biotinylated BTK WT enzym (Carna Biosciences, cat. no. 08-480-20N), BTK C481 S (Carna Biosciences, cat. no. OS- 417-20N), BTK T474I (Carna Biosciences, cat. no. 08-419-20N) were immobilized on a streptavidin-coated chip to a level of about 8000 resonance units (RU) using Biacore buffer (50 mM Tris pH 7.5, 0.05 % (v/v) Tween-20, 150 mM NaCI and 5 mM MgCI2) + 1 mM TCEP. 10 Remaining streptavidin was blocked with biocytin. Immobilization was performed at 4°C. Subsequent assay steps were conducted at 22°C. After changing buffer to Biacore buffer with 1 % (v/v) dimethylsulfoxide (DMSO), a pre-run was performed for a period of at least 30 min at a flow rate of 30 pl/min to obtain a stable surface. The kinetic constants of the compounds were determined with single cycle kinetics with five consecutive injections with an increasing 15 compound concentration with ranges of 3.16 - 316 nM. Experiments were performed with an association time of 100 s per concentration and a dissociation time of 1200 s, except for compounds with a long target residence time, such as irreversible inhibitors, where dissociation time was increased. To circumvent problems of mass transport limitation, a flow rate of 30 pl/min was used. A blank run with the same conditions was performed before the compound was 20 injected. The SPR sensorgrams were analyzed with Biacore Evaluation Software by using a method of double referencing. First the reference channel was subtracted from the channel containing immobilized protein. Subsequently, the reference curve obtained with buffer injections was subtracted. The resulting curve was fitted with a 1 :1 binding model. Compounds that bound according to an induced fit model were fitted with a two-state reaction model. The kinetic 25 constants (ka, kd, KD) of duplicates were geometrically averaged. Target residence time (T) for the 1 :1 binding model was calculated from the dissociation constant k^with the formula T = 1 / d. Target residence for an induced fit model was calculated as described (Tummino and Copeland, 2008).
Generation of human wt-BTK, BTK C481 S, BTK T474I, BTK T474I/C481S and BTK M437R expressing GripTite 293 MSR cells.
GripTite 293 MSR cells (ThermoScientific, cat. no. R79507), here after referred to as 293 cells, were cultured in DMEM/F-12, GlutaMAX™ supplement medium (ThermoScientific cat. no. 10565018) supplemented with 1% Penicilline/Streptomycine (ThermoScientific cat. no. 15140122 ), 10% fetal bovine serum (Biowest, cat. no. S1810-500), 1x MEM Non-Essential
Amino Acids (ThermoScientific cat. no. 11140050) and 50 pg/ml Geneticin (ThermoScientific cat. no. 10131035). Cells were transfected, with pEF6/V5-HisB (ThermoScientific cat. no. V96120) expression vectors containing either full-length wt-BTK (canonical sequence NM_000061 ), BTK C481S, BTK T474I, BTK T474I/C481S and BTK M437R (BaseClear) using Lipofectamine™ 3000 (Invitrogen). Because the stop codon at the end of the coding sequence of BTK has been conserved, the His-tag present in the expression vector is not being used. Immediately after transfection, cells were cultured in medium without Geneticin, and after 24 hours 50 pg/ml geneticin was added. 48 hours after transfection, medium was removed and replaced by medium containing both geneticin (50 pg/ml) and blasticidin (10 pg/ml) S HCI (ThermoScientific, cat. no. A11 13903). The resulting transfectants were cultured under blasticidin selection pressure for 18-35 passages to obtain stable wt-BTK, BTK C481 S, BTK T474I, BTK T474I/C481 S and BTK M437R expressing cell pools. Stability of the (mutant) BTK expression in the 293 cell pools (wt-BTK pool B3, BTK C481 S pool B5, BTK T474I pool C2, BTK T474I/C481S pool B3 and BTK M437R) was confirmed by poly-acrylamide gel electrophoresis followed by Western blotting. Non-transfected 293 cells do not express BTK.
SDS-PAGE and Western blotting wt-BTK, BTK C481S, BTK T474I, BTK T474I/C481S and BTK M437R expressing 293 cells were lysed in lysis buffer supplemented with protease inhibitors (ThermoScientific, cat. no. 78429) and phosphatase inhibitors (ThermoScientific, cat. no. 78426). Cleared lysates were diluted in NuPAGE™ LDS Sample Buffer (ThermoScientific, cat. no. NP0007) supplemented with NuPAGE™ Sample Reducing Agent (ThermoScientific, cat. no. NP0009) and denatured for 5 minutes at 95°C. The samples were, together with a pre-stained protein ladder (ThermoScientific, cat. no. 26616 or 26619), separated on 4-12% Bis-Tris polyacrylamide gels (ThermoScientific, cat. no. NP0324BOX). Separated protein was subsequently transferred, in methanol free transfer buffer (ThermoScientific, cat. no. 35045), to a nitrocellulose membrane (ThermoScientific, cat. no. 88018). Blots were first immunologically stained with phospho-BTK (Y223) rabbit mAb (Cell Signaling, cat. no. 5082S), and beta-actin rabbit mAb (Cell Signalling, cat. no. 4967S), followed by peroxidase conjugated goat anti-rabbit IgG (Cell Signalling, cat. no. 7074S). After imaging, antibodies were removed using Western-blot stripping buffer (ThermoScientific cat. no. 46428) and staining was repeated with a (total-)BTK mouse mAb (Cell Signalling, cat. no. 56044S), followed by peroxidase conjugated horse anti-mouse IgG (Cell Signalling, cat. no. 7076S). For all blots the colour development was performed with ECL horseradish peroxidase substrate (BioRad, cat. no. 170-5060).
Compound preparation
All compounds, obtained from commercial suppliers or synthesized in house, were dissolved in 100 % DMSO (Acres, cat. no. 127790010), and stored at room temperature. At the start of the experiments the compound stock was diluted in 3.16-fold dilution steps in 100% DMSO to obtain
a 7-point dilution series, followed by further dilution in culture medium or 20 mM Hepes. Final test concentrations in the BTK inhibition assay were 1 nM to 1000 nM in 0.3% DMSO.
Cell based BTK inhibition wt-BTK, BTK C481S, BTK T474I, and BTK T474I/C481 S and BTK M437R expressing 293 cells were growing exponentially when seeded. Cells were seeded in culture medium without Blasticidin, at a density of 30.000 cells/well in 96-well plates (90 pL/well) (Greiner Bio-one, cat. no. 655182), and placed in a 37 °C CO2-incubator. Between 16 and 24 hours after seeding, 20 mM Hepes with compound or DMSO was added to the cells (10 pL). Two hours after addition, medium with compound was completely aspirated and cells were lysed in lysis buffer containing protease inhibitors and phosphatase inhibitors. Sample preparation, SDS-PAGE and Western blotting was performed as described under ‘SDS-PAGE and Western blotting’.
Cell based wash-out
WT-BTK, BTK C481S, BTK T474I, BTK T474I/C481S and BTK M437R expressing 293 cells were growing exponentially when seeded, in culture medium, without blasticidin, at a density of 2.000.000 cells/4mL, in 25 cm2 cell culture flasks (Greiner Bio-one, cat. no. 690175). Flasks were placed in a 37 °C CO2-incubator for 16-24 hours, followed by addition of compound (in medium). Two hours after addition of the inhibitors (Oh) cells are either collected or washed twice with 5 mL culture medium. After washing, 5 mL culture medium (without blasticidin), was added and cells were incubated at 37°C in a CO2 incubator for 0.5, 1 , 2, 4, 6, or 24 hours before being collected. Cells were harvested using 0.25% trypsin (ThermoScientific, cat. no. 25200056). After harvesting, cells pellets were washed twice with PBS (ThermoScientific, cat. no. 14190-094), snap-frozen, and stored at -80°C. Sample preparation, SDS-PAGE and Western blotting was performed as described under ‘SDS-PAGE and Western blotting’.
Results
Proliferation assay of single compounds (monotherapy) in wt-BTK TMD8 cells (Figure 2)
Viability of Compound A and other compounds in wt-BTK TMD8 cells. All compounds inhibit proliferation with <10 nM IC50S and >90% efficacy, nemtabrutinib excluded. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability at the highest tested concentration from vehicle treated cells. Dotted line indicates the cell population at the start of the experiment (t=0).
Table T. Proliferation data for wt-BTK TMD8 cells
Proliferation assay of single compounds (monotherapy) in BTK C481S TMD8 cells. (Figure
3)
Viability of Compound A and other compounds in BTK C481 S TMD8 cells. Compound A, pirtobrutinib and nemtabrutinib inhibit proliferation. Ibrutinib and acalabrutinib do not inhibit proliferation at the tested concentrations. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability at the highest tested concentration from vehicle treated cells. Dotted line indicates the cell population at the start of the experiment (t=0).
Table 2 Proliferation data for BTK C481 S TMD8 cells
Proliferation assay of single compounds (monotherapy) in BTK T474I TMD8 cells. (Figure 4)
Viability of Compound A and other compounds in BTK T474I TMD8 cells. Compound A, ibrutinib and acalabrutinib inhibit proliferation. Pirtobrutinib and nemtabrutinib do not inhibit proliferation at the tested concentrations. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability at the highest tested concentration from vehicle treated cells. Dotted line indicates the cell population at the start of the experiment (t=0).
Table 3: Proliferation data for BTK T474I TMD8 cells
Proliferation assay of single compounds (monotherapy) in BTK T474I/C481S TMD8 cells.
(Figure 5)
Viability of Compound A and other compounds in BTK T474I/C481 S TMD8 cells. Only Compound A inhibits proliferation. All other compounds are unable to inhibit proliferation at the tested concentrations. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability at the highest tested concentration from vehicle treated cells. Dotted line indicates the cell population at the start of the experiment (t=0).
Table 4: Proliferation data for BTK T474I/C481 S TMD8 cells
Proliferation assay of single compounds (monotherapy) in BTK V416L TMD8 cells. (Figure 6)
Viability of Compound A and other compounds in BTK V416L TMD8 cells. Compound A, ibrutinib and acalabrutinib inhibit proliferation. Pirtobrutinib and nemtabrutinib do not inhibit proliferation at the tested concentrations. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability at the highest tested concentration from vehicle treated cells. Dotted line indicates the cell population at the start of the experiment (t=0).
Proliferation assay of single compounds (monotherapy) in BTK L528W TMD8 cells. (Figure 7)
Viability of Compound A and other compounds in BTK L528W TMD8 cells. Compound A and acalabrutinib inhibit the proliferation. Ibrutinib and pirtobrutinib show low efficacy and nemtabrutinib does not inhibit proliferation at the tested concentrations. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability at the highest tested concentration from vehicle treated cells. Dotted line indicates the cell population at the start of the experiment (t=0).
Table 6: Proliferation data for BTK L528W TMD8 cells
Proliferation assay of single compounds (monotherapy) in a mixture of BTK C481S TMD8 cells and BTK V416L TMD8 cells (Figure 8)
Viability of compound A, ibrutinib, acalabrutinib and pirtobrutinib in a 1 :1 mixture of BTK C481 S TMD8 cells + BTK V416L TMD8 cells after 96h incubation. Graphs represent efficacy of compounds used in the standard of care. Compound A has a higher efficacy than ibrutinib, acalabrutinib and pirtobrutinib. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability of the highest tested concentration from vehicle treated cells. The dotted line indicates the cell population at the start of the experiment (t=0).
Table 7: Proliferation data for compounds in a mixture of BTK C481 S TMD8 cells + BTK V416L TMD8 cells.
Proliferation assay of single compounds (monotherapy) in a mixture of BTK C481S TMD8 cells and BTK T474I TMD8 cells (Figure 9)
Viability of Compound A, ibrutinib, acalabrutinib and pirtobrutinib in a 1 :1 mixture of BTK C481 S TMD8 cells + BTK T474I TMD8 cells after 96h incubation. Graphs of ibrutinib, acalabrutinib and pirtobrutinib represent efficacies of the standard of care. Compound A has a higher efficacy than ibrutinib, acalabrutinib and pirtobrutinib. Vehicle treated cells represent 100% viability. Efficacy was calculated by subtracting % viability of the highest tested concentration from vehicle treated cells. The dotted line indicates the cell population at the start of the experiment (t=0).
Table 8 Proliferation data for compounds in a mixture of BTK C481 S TMD8 cells + BTK T474I TMD8 cells.
Cell based washout (Western Blot)
Wash-out Western blot results of wt-BTK expressing 293 cells. Cells were incubated with 1000 nM of compound A or 1000 nM of pirtobrutinib in cell culture medium without blasticidin. Compound containing medium was removed after 2h hours and cells were washed twice before being replaced by medium without compound. Cells were harvested after 0, 0.5, 1 , 2, 4, 6 and 24 hours after medium replacement. Control cells (no compound) were incubated for two hours with 0.01 % DMSO (D) and harvested at 0 hours (Oh), which is at the start of the wash-out, and 24 hours (24h). The level of phosphorylated BTK (pBTK Y223) was determined on Western blot as shown in Figure 10A. Expression of phosphorylated BTK was quantified relative to expression of phosphorylated BTK after incubation with 0.01 % DMSO. Expression was normalized using beta-actin as shown in Figure 10B.
Wash-out Western blot results of BTK C481 S expressing 293 cells. Cells were incubated with 1000 nM of compound A or 1000 nM of pirtobrutinib in cell culture medium without blasticidin. Compound containing medium was removed after 2h hours and cells were washed twice before being replaced by medium without compound. Cells were harvested after 0, 0.5, 1 , 2, 4, 6 and 24 hours after medium replacement. Control cells (no compound) were incubated for two hours with 0.01 % DMSO (D) and harvested at 0 hours (Oh), which is at the start of the wash-out, and 24 hours (24h). The level of phosphorylated BTK (pBTK Y223) was determined on Western blot as shown in Figure 11A. Expression of phosphorylated BTK was quantified relative to expression of phosphorylated BTK after incubation with 0.01 % DMSO. Expression was normalized using beta-actin as shown in Figure 11 B.
Wash-out Western blot results of BTK T474I expressing 293 cells. Cells were incubated with 1000 nM of compound A or 1000 nM of pirtobrutinib in cell culture medium without blasticidin. Compound containing medium was removed after 2h hours and cells were washed twice before being replaced by medium without compound. Cells were harvested after 0, 0.5, 1 , 2, 4, 6 and 24 hours after medium replacement. Control cells (no compound) were incubated for two hours with 0.01 % DMSO (D) and harvested at 0 hours (Oh), which is at the start of the wash-out, and 24 hours (24h). The level of phosphorylated BTK (pBTK Y223) was determined on Western blot as shown in Figure 12A. Expression of phosphorylated BTK was quantified relative to expression of phosphorylated BTK after incubation with 0.01 % DMSO. Expression was normalized using beta-actin as shown in Figure 12B.
Wash-out Western blot results of BTK T474I/C481 S expressing 293 cells. Cells were incubated with 1000 nM of compound A or 1000 nM of pirtobrutinib in cell culture medium without blasticidin. Compound containing medium was removed after 2h hours and cells were washed twice before being replaced by medium without compound. Cells were harvested after 0, 0.5, 1 ,
2, 4, 6 and 24 hours after medium replacement. Control cells (no compound) were incubated for two hours with 0.01 % DMSO (D) and harvested at 0 hours (Oh), which is at the start of the washout, and 24 hours (24h). The level of phosphorylated BTK (pBTK Y223) was determined on Western blot as shown in Figure 13A. Expression of phosphorylated BTK was quantified relative to expression of phosphorylated BTK after incubation with 0.01 % DMSO. Expression was normalized using beta-actin as shown in Figure 13B.
Wash-out Western blot results of BTK M437R expressing 293 cells. Cells were incubated with 1000 nM of compound A or 1000 nM of pirtobrutinib in cell culture medium without blasticidin. Compound containing medium was removed after 2h hours and cells were washed twice before being replaced by medium without compound. Cells were harvested after 0, 0.5, 1 , 2, 4, 6 and 24 hours after medium replacement. Control cells (no compound) were incubated for two hours with 0.01 % DMSO (D) and harvested at 0 hours (Oh), which is at the start of the wash-out, and 24 hours (24h). The level of phosphorylated BTK (pBTK Y223) was determined on Western blot as shown in Figure 14A. Expression of phosphorylated BTK was quantified relative to expression of phosphorylated BTK after incubation with 0.01 % DMSO. Expression was normalized using beta-actin as shown in Figure 14B.
It is clear from the wash-out experiments that pirtobrutinib does not prohibit or reduce the level of phosphorylated BTK (pBTK Y223), while compound A prohibits or reduces the level of phosphorylated BTK (pBTK Y223) for at least 6 hours up till at least 24 hours, even for cell lines expressing mutant variations of BTK. This shows that the target residence of the compounds according to the invention is still substantial to complete during said period after removing the solution of compounds.
Cell based Dose Response Curves (Western Blot)
Western blot protein expression results of wt-BTK, BTK C481 S, BTK T474I, and BTK T474I/C481 S expressing 293 cells incubated with a dose response range of compound A (Fig. 15A), pirtobrutinib (Fig. 15B), ibrutinib (Fig.15C), acalabrutinib (Fig. 15D) and nemtabrutinib (Fig. 15E). DMSO was used as a vehicle control. 2 hours after addition of compound, cells were lysed and the level of BTK phosphorylation (pBTK Y223) was determined on Western blot. Beta-actin (ACTB) and BTK (total BTK) expression levels were used as controls.
Western blot protein expression results of BTK M437R expressing 293 cells incubated with a dose response range of compound A, pirtobrutinib, ibrutinib, acalabrutinib, and nemtabrutinib (Fig. 15F). DMSO was used as a vehicle control. 2 hours after addition of compound, cells were lysed and the level of BTK phosphorylation (pBTK Y223) was determined on Western blot. Betaactin (ACTB) and BTK (total BTK) expression levels were used as controls.
Surface plasma resonance
Target residence times for compound A and reference reversible inhibitors are given in Table 9 below.
Table 9: KD and Target residence times (T (h)) for BTK inhibitors on wt-BTK, BTK C481S, BTK
REFERENCES
• Attwood et al. (2021) “Trends in kinase drug discovery targets, indications and inhibitor design.” Nat Rev Drug Discov 20, pages 839-861 (2021).
• Al-Lazikani B, Banerji U, Workman P (2012) Combinatorial drug therapy for cancer in the post-genomic era. Nature Biotech 30: 679-682.
• Yap TA, Omlin A, de Bono JS (2013) Development of therapeutic combinations targeting major cancer signaling pathways. J Clinical Invest 20: 1592-1605.
• Li F, Zhao C, Wang L (2014) Molecular-targeted agents combination therapy for cancer: developments and potentials. Int J Cancer 134: 1257-1269.
• Borisy AA, Elliott PJ, Hurst NW, Lee MS, Lehar J, Price ER, et al. (2003) Systematic discovery of multicomponent therapeutics. Proc Natl Acad Sci USA; 100: 7977-7982.
• Cokol M, Chua HN, Tasan M, Mutlu B, Weinstein ZB, Suzuki Y, et al. (2011) Systematic exploration of synergistic drug pairs. Molecular systems biology 7: 544.
• Mathews Griner LA, Guha R, Shinn P, Young RM, Keller JM, Liu D, et al. (2014) High throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells. Proc Natl Acad Sci USA 111 : 2349-2354.
• Zhao L, Wientjes MG, Au JLS (2004) Evaluation of combination chemotherapy: integration of nonlinear regression, curve shift, isobologram and combination index analyses. Clin Cane Res 10: 7994-8004.
• Chou, T-C (2010) Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 70: 440-446.
• Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. (2012) Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483: 570-575.
• Straetemans R, O’Brien T, Wouters L, van Dun J, Janicot M, et al. (2005) Design and analysis of drug combination experiments. Biometrical J 47: 299-308.
• Nakhoda, S., Vistarop, A., & Wang, Y. L. (2022). Resistance to Bruton tyrosine kinase inhibition in chronic lymphocytic leukaemia and non-Hodgkin lymphoma. British Journal of Haematology, 200(2), 137-149.
• R. Barouch-Bentov and K. Sauer (2011) Mechanisms of drug resistance in kinases. Exp. Opin. Invest. Drugs, 20:153-208.
• Woyach, J. A., Ruppert, A. S., Guinn, D., Lehman, A., Blachly, J. S., Lozanski, A., Heerema, N. A., Zhao, W., Coleman, J. T., Jones, D. B., Abruzzo, L. V., Gordon, A., Mantel, R., Smith, L., McWhorter, S., Davis, M. J., Doong, T. J., Ny, F., Lucas, M., . . . Byrd, J. C. (2017). BTKC481 S-Mediated Resistance to Ibrutinib in Chronic Lymphocytic Leukemia. Journal of Clinical Oncology, 35(13), 1437-1443.
• Reiff, S. D., Mantel, R., Smith, L., Greene, J. E., Muhowski, E. M., Fabian, C. A., Goettl, V. M., Tran, M. T., Harrington, B. K., Rogers, K. A., Awan, F. T., Maddocks, K. J., Grever, M. R., Lehman, A., Sampath, D., Lapalombella, R., Eathiraj, S., Abbadessa, G., Schwartz, B. S., . . . Woyach, J. A. (2018). The BTK Inhibitor ARQ 531 Targets Ibrutinib-Resistant CLL and Richter Transformation. Cancer Discovery, 8(10), 1300- 1315.
• Crawford, J., Johnson, A. R., Misner, D., Belmont, L. D., Castanedo, G., Choy, R., Coraggio, M., Dong, L., Eigenbrot, C., Erickson, R. J., Ghilardi, N., Hau, J., Katewa, A., Kohli, P. B., Lee, W., Lubach, J. W., McKenzie, B. S., Ortwine, D. F., Schutt, L., . . . Young, W. B. (2018). Discovery of GDC-0853: A Potent, Selective, and Noncovalent Bruton’s Tyrosine Kinase Inhibitor in Early Clinical Development. Journal of Medicinal Chemistry, 61 (6), 2227-2245.
• Wang, E. W., Mi, X., Thompson, M. C., Montoya, S., Notti, R. Q., Afaghani, J., Durham, B. H., Penson, A. V., Witkowski, M. T., Lu, S. X., Bourcier, J., Hogg, S. J., Erickson, C., Cui, D., Cho, H., Singer, M. B., Totiger, T. M., Chaudhry, S., Geyer, M. A., . . . Abdel-Wahab, O. (2022). Mechanisms of Resistance to Noncovalent Bruton’s Tyrosine Kinase Inhibitors. The New England Journal of Medicine, 386(8), 735-743.
• Watterson, S. H., De Lucca, G. V., Shi, Q., Langevine, C. M., Liu, Q., Batt, D. G., Bertrand, M. B., Gong, H., Dai, J., Yip, S., Li, P., Sun, D., Wu, D., Wang, C., Zhang, Y., Traeger, S. C., Pattoli, M. A., Skala, S., Cheng, L., . . . Tino, J. A. (2016). Discovery of 6-Fluoro-5-(R)-(3-(S)-(8-fluoro-1-methyl-2,4-dioxo-1 ,2-dihydroquinazolin-3(4H)-yl)-2- methylphenyl)-2-(S)-(2-hydroxypropan-2-yl)-2,3,4,9-tetrahydro-1 H-carbazole-8- carboxamide (BMS-986142): A Reversible Inhibitor of Bruton’s Tyrosine Kinase (BTK) Conformationally Constrained by Two Locked Atropisomers. Journal of Medicinal Chemistry, 59(19), 9173-9200.
• Liu, Q., Yin, X., Languino, L. R., & Altieri, D. C. (2018). Evaluation of Drug Combination Effect Using a Bliss Independence Dose-Response Surface Model. Statistics in Biopharmaceutical Research, 10(2), 112-122.
• Wang, X., Kokabee, L., Kokabee, M., & Conklin, D. S. (2021). Bruton’s Tyrosine Kinase and Its Isoforms in Cancer. Frontiers in Cell and Developmental Biology, 9.
• Kokabee, L., Wang, X., Sevinsky, C. J., Wang, W., Cheu, L., Chittur, S. V., Karimipoor, M., Tenniswood, M., & Conklin, D. S. (2015). Bruton’s tyrosine kinase is a potential therapeutic target in prostate cancer. Cancer Biology & Therapy, 16(11), 1604-1615.
• Wang, X., Wong, J. W. H., Sevinsky, C. J., Kokabee, L., Khan, F. M., Sun, Y., & Conklin, D. S. (2016). Bruton’s Tyrosine Kinase Inhibitors Prevent Therapeutic Escape in Breast Cancer Cells. Molecular Cancer Therapeutics, 15(9), 2198-2208.
• Hopkins, B., Bame, E., Bajrami, B., Black, C. A., Bohnert, T., Boiselle, C., Burdette, D., Burns, J. C., Delva, L., Donaldson, D., Grater, R., Gu, C., Hoemberger, M., Johnson, J.
R., Kapadnis, S., King, K., Lulla, M., Ma, B., Marx, I. E., . . . Lyssikatos, J. P. (2021). Discovery and Preclinical Characterization of BIIB091 , a Reversible, Selective BTK Inhibitor for the Treatment of Multiple Sclerosis. Journal of Medicinal Chemistry, 65(2), 1206-1224.
• Thieme, E., Liu, T., Bruss, N., Roleder, C., Lam, V., Wang, X., Nechiporuk, T., Shouse, G., Danilova, O. V., Bottomly, D., McWeeney, S. K., Tyner, J. W., Kurtz, S. E., & Danilov, A. V. (2022). Dual BTK/SYK inhibition with CG-806 (luxeptinib) disrupts B-cell receptor and Bcl-2 signaling networks in mantle cell lymphoma. Cell Death and Disease, 13(3).
• Kawahata, W., Asami, T., Kiyoi, T., Irie, T., Taniguchi, H., Asamitsu, Y., Inoue, T., Miyake, T., & Sawa, M. (2018). Design and Synthesis of Novel Amino-triazine Analogues as Selective Bruton’s Tyrosine Kinase Inhibitors for Treatment of Rheumatoid Arthritis. Journal of Medicinal Chemistry, 61 (19), 8917-8933.
• Kawahata, W., Asami, T., Kiyoi, T., Irie, T., Kashimoto, S., Furuichi, H., & Sawa, M. (2021). Discovery of AS-1763: A Potent, Selective, Noncovalent, and Orally Available Inhibitor of Bruton’s Tyrosine Kinase. Journal of Medicinal Chemistry, 64(19), 14129— 14141.
• Reiff, S. D., Muhowski, E. M., Guinn, D., Lehman, A., Fabian, C. A., Cheney, C., Mantel, R., Smith, L., Johnson, A. J., Young, W. B., Johnson, A. R., Liu, L., Byrd, J. C., & Woyach, J. A. (2018). Noncovalent inhibition of C481 S Bruton tyrosine kinase by GDC-0853: a new treatment strategy for ibrutinib-resistant CLL. Blood, 132(10), 1039- 1049.
• Bodor, C., Kotmayer, L., Laszlo, T., Takacs, F., Barna, G., Kiss, R., Sebestyen, E., Nagy, T., Hegyi, L., Mikala, G., Fekete, S. P., Farkas, P., Balogh, A., Masszi, T., Demeter, J., Weisinger, J., Alizadeh, H., Kajtar, B., Kohl, Z., . . . Alpar, D. (2021). Screening and monitoring of the BTK C481S mutation in a real-world cohort of patients with relapsed/refractory chronic lymphocytic leukaemia during ibrutinib therapy. British Journal of Haematology, 194(2), 355-364.
• Wei, L., Su, Y„ Lin, C. M., Chao, T. Y., Huang, S., Huynh, T. D„ Jan, H. J., Whang- Peng, J., Chiou, J. F., Wu, A. T., & Hsiao, M. (2016). Preclinical investigation of ibrutinib, a Bruton’s kinase tyrosine (Btk) inhibitor, in suppressing glioma tumorigenesis and stem cell phenotypes. Oncotarget, 7(43), 69961-69975.
Claims
Claims
1 . A reversible BTK inhibitor for use in a method for treating a subject diagnosed with or at risk of a recurrent or refractory from of a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has previously been treated with a BTK inhibitor, preferably an irreversible BTK inhibitor, and wherein the method comprises administering to said subject an effective amount of said reversible BTK inhibitor , which is a compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is selected from the group consisting of:
W is an aryl group having 6-10 carbon or a heteroaryl group having 1-5 carbon; wherein any said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;
V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R1v)-NH-C(O)-, -CH(R1v)- ;
R1v is hydrogen or (1 -2C)alkyl;
U is an aryl group having 6-10 carbon or an heteroaryl group having 1-5 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl, alkoxy, cycloalkyl and heterocycloalkyl group is optionally and independently substituted with one, two or three halogen; wherein R2 is of Formula (I l-a) to (ll-f) selected from the group consisting of:
(Formula ll-d) (Formula ll-e) (Formula ll-f) wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein Xi, X2 and X3 are independently selected from CH2, -CH2CH2-, O, N and a direct bond; wherein any of the cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with one or more substituents selected from halogen, hydroxy, (1 - 3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R3 and R4 together represent a linker having Formula (111-1 to HI-40) selected from the group consisting of:
(HI-34) (HI-35) (HI-36) (HI-37) (HI-38) (HI-39) (HI-40)
whereby the
* marks the position of R3 in any one of Formula l-a to l-h, and whereby the
marks the position of R4 in any one of Formula I l-a to I l-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CDs, (1 -4C)alkyl, (1 - 5C)alkoxy, (3-6C)cycloalkyl, (3-6C)cycloalkoxy and (1-6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.
2. A reversible BTK inhibitor for use in a method according to claim 1 , wherein the subject has received at least one of ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, spebrutinib, branebrutinib, evobrutinib, remibrutinib, tolebrutinib, orelabrutinib, elsubrutinib, edralbrutinib, ACP-5862, preferably at least one of ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, before recurrence or relapse of the hyperproliferative disease.
3. A reversible BTK inhibitor for use in a method for treating a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the hyperproliferative disease, preferably an irreversible BTK inhibitor, the method comprising the steps:
(a) monitoring the subject, preferably at predetermined intervals of time, over the course of the therapy for treatment of the hyperproliferative disease to determine whether the subject has a mutation in an endogenous gene encoding BTK that results in resistance or provides a risk of resistance of the subject to the therapy, preferably resistance of the subject to the irreversible BTK inhibitor, preferably the subject has a mutation in an endogenous gene encoding BTK that results in expression of a BTK protein with a modification at an amino acid position corresponding to amino acid position 481 of the amino acid sequence according to SEQ ID NO: I; and
(b) if the subject has shown said mutation, administering to the subject a reversible BTK inhibitor, in particular if the subject has the BTK modification corresponding to amino acid position 481 identified in step (a), wherein the reversible BTK inhibitor is a compound of any one of Formula (l-a) to (l-h) as defined in claim 1 .
4. A reversible BTK inhibitor for use in a method for treating a subject having a hyperproliferative disease, preferably a B-cell hematological malignancy, which subject receives or has received a Bruton’s tyrosine kinase (BTK) inhibitor for treatment of the
hyperproliferative disease, preferably an irreversible BTK inhibitor, wherein the method comprises: optionally stopping the administration of the irreversible BTK inhibitor, and administering to said subject an effective amount of a reversible BTK inhibitor, which is a compound of any one of Formula (l-a) to (l-h) as defined in claim 1 .
5. Reversible BTK inhibitor for use in a method of any one of claims 1-4, wherein the irreversible BTK inhibitor is selected from the group consisting of: ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, spebrutinib, branebrutinib, evobrutinib, remibrutinib, tolebrutinib, orelabrutinib, elsubrutinib, edralbrutinib, ACP-5862, and pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
6. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the subject has been administered an irreversible BTK inhibitor for treatment of the hyperproliferative disease, preferably the B-cell hematological malignancy, preferably before recurrence or relapse of the hyperproliferative disease.
7. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the protein modification is C481S.
8. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 of the amino acid sequence according to SEQ ID NO: I, wherein the modification is a substitution of cysteine to an amino acid selected from leucine, isoleucine, valine, alanine, glycine, methionine, serine, threonine, phenylalanine, tryptophan, lysine, arginine, histidine, proline, tyrosine, asparagine, glutamine, aspartic acid and glutamic acid at amino acid position 481 of the BTK protein.
9. Reversible BTK inhibitor for use in a method of claim 8, wherein the modification is a substitution of cysteine to serine at amino acid position 481 of the BTK protein.
10. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the reversible BTK inhibitor is a compound according to Formula (l-a) or (l-b) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound comprises a bicyclic scaffold selected from:
(Formula l-a) (Formula l-b)
Wherein R1 is any one of:
wherein R2w is selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any said alkyl or alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R3u is selected from hydrogen, halogen, cyano, (1 -4C)alkyl, (1 -5C)alkoxy, (3- 6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R2 is selected from the group consisting of:
(Formula Il-a4) (Formula Il-b3) (Formula Il-c3)
(Formula ll-d) (Formula ll-f) (Formula Il-a5) (Formula Il-b4) wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with hydroxy, methyl, acetyl or methoxy;
wherein R3 and R4 together represent a linker having Formula selected from the group consisting of:
whereby the ”” * marks the position of R3 in any one of Formula l-a to l-b, and whereby the ■~ ~ marks the position of R4 in any one of Formula I l-a to I l-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CDs, (1 -4C)alkyl, (1 - 5C)alkoxy, (3-6C)cycloalkyl, (3-6C)cycloalkoxy and (1-6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.
11 . Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the reversible BTK inhibitor is a compound according to Formula A:
(A), or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
12. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the irreversible BTK inhibitor is selected from the group consisting of: ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, and pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
13. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the irreversible BTK inhibitor is ibrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
14. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the irreversible BTK inhibitor is acalabrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
15. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the irreversible BTK inhibitor is zanubrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
16. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the irreversible BTK inhibitor is tirabrutinib, or pharmaceutically acceptable salts, cocrystals, hydrates, solvates, and prodrugs thereof.
17. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the subject is identified as having one or more mutations in a gene that results in
expression of a BTK protein with a modification at one or more amino acid positions selected from the group consisting of: 416, 428, 437, 474, 481 and 528.
18. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 416 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is V416L.
19. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 428 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is A428D.
20. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 437 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is M437R.
21 . Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 474 of the amino acid sequence set according to SEQ ID NO: I, more preferably wherein the mutant modification is T474I.
22. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 528 of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is L528W.
23. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the subject is monitored at predetermined intervals of time over the course of the prior therapy for treatment of the hyperproliferative disease, wherein the predetermined interval of time is every week, every 2 weeks, every 3 weeks, every month, every 2 months, every 3 months, every 4 months, every 5 months, every 6 months, every 7 months, every 8 months, every 9 months, every 10 months, every 11 months, every year following the first administration of the irreversible BTK inhibitor.
24. Reversible BTK inhibitor for use in a method of any one of preceding claims, wherein the irreversible BTK inhibitor is administered during the prior therapy in an amount in the range of 70 - 750 mg / day and / or wherein the irreversible BTK inhibitor is administered using one or more unit doses having an amount in the range of 70 - 750 mg / unit dose.
25. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the irreversible BTK inhibitor is administered during the prior therapy daily at a dose selected from the group consisting of 70 mg, 100 mg, 140 mg, 160 mg, 200 mg, 280 mg, 320 mg, 420 mg, 480 mg, 560 mg per day.
26. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the irreversible BTK inhibitor is administered during the prior therapy twice daily at a dose of 80 mg, 100 mg or 160 mg.
27. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the hyperproliferative disease is a B-cell hematological malignancy.
28. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the B cell hematological malignancy is any one of chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), diffuse large B-cell lymphoma (DLBCL), activated B-cell diffuse large B-cell lymphoma (ABC-DLBCL), germinal center diffuse large B-cell lymphoma (GCB DLBCL), primary mediastinal B-cell lymphoma (PMBL), non-Hodgkin lymphoma, Burkitt’s lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, precursor B-cell acute lymphoblastic leukemia, hairy cell leukemia, mantle cell lymphoma, B-cell prolymphocytic leukemia, lym- phoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis.
29. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the B cell hematological malignancy is any one of mantle cell lymphoma (MCL), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), diffuse large B-cell lymphoma (DLBCL), Waldenstrom macroglobulinemia (WM), follicular lymphoma (FL) and marginal zone lymphoma (MZL).
30. Reversible BTK inhibitor for use in a method of any one of the preceding claims, wherein the B-cell malignancy is Mantle cell lymphoma (MCL) or chronic lymphocytic leukemia (CLL). 31 . Reversible BTK inhibitor for use in a method of any one of the claim 27 - 30, wherein the B-cell hematological malignancy of the subject is relapsed or refractory, preferably wherein CLL/SLL is relapsed or refractory.
32. Reversible BTK inhibitor for use in a method of any one of the claim 27 - 31 , wherein the B-cell hematological malignancy of the subject is relapsed or refractory and the subject is identified as having a mutation in a gene that results in expression of a BTK protein with a modification at amino acid position 481 , of the amino acid sequence according to SEQ ID NO: I, more preferably wherein the mutant modification is C481S.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EPPCT/EP2023/064842 | 2023-06-02 | ||
| EP2023064842 | 2023-06-02 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024246287A1 true WO2024246287A1 (en) | 2024-12-05 |
Family
ID=86776512
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/065039 Pending WO2024246287A1 (en) | 2023-06-02 | 2024-05-31 | Medical use of a macrocyclic reversible btk inhibitor |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024246287A1 (en) |
Citations (21)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2013010380A1 (en) | 2011-07-19 | 2013-01-24 | Merck Sharp & Dohme Corp. | Btk inhibitors |
| WO2013067274A1 (en) | 2011-11-03 | 2013-05-10 | Genentech, Inc. | Heteroaryl pyridone and aza-pyridone compounds as inhibitors of btk activity |
| WO2013185084A1 (en) | 2012-06-08 | 2013-12-12 | Biogen Idec Ma Inc. | Pyrimidinyl tyrosine kinase inhibitors |
| WO2014018567A1 (en) * | 2012-07-24 | 2014-01-30 | Pharmacyclics, Inc. | Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk) |
| WO2016109222A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Tertiary alcohol imidazopyrazine btk inhibitors |
| WO2016106626A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Imidazopyrazine analogs with 3-tertiary carbon substitutions as btk inhibitors |
| WO2016106628A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Btk inhibitors |
| WO2016106625A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Btk inhibitors |
| WO2016106623A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Benzamide imidazopyrazine btk inhibitors |
| WO2016161571A1 (en) | 2015-04-08 | 2016-10-13 | Merck Sharp & Dohme Corp. | Indazole and azaindazole btk inhibitors |
| WO2016161570A1 (en) | 2015-04-08 | 2016-10-13 | Merck Sharp & Dohme Corp. | Azacarbazole btk inhibitors |
| WO2017046604A1 (en) | 2015-09-16 | 2017-03-23 | Redx Pharma Plc | Pyrazolopyrimidine derivatives as btk inhibitors for the treatment of cancer |
| WO2017103611A1 (en) | 2015-12-16 | 2017-06-22 | Redx Pharma Plc | Compounds useful as kinase inhibitors |
| WO2017111787A1 (en) | 2015-12-23 | 2017-06-29 | Arqule, Inc. | Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof |
| WO2018097234A1 (en) | 2016-11-25 | 2018-05-31 | カルナバイオサイエンス株式会社 | Novel oxoisoquinoline derivative |
| WO2020015735A1 (en) | 2018-07-20 | 2020-01-23 | 正大天晴药业集团股份有限公司 | Bruton tyrosine kinase inhibitors |
| WO2020043638A1 (en) | 2018-08-28 | 2020-03-05 | Merck Patent Gmbh | Fused imidazopyridines as reversible inhibitors of bruton's tyrosine kinase (btk) |
| WO2020239124A1 (en) | 2019-05-31 | 2020-12-03 | Fochon Pharmaceuticals, Ltd. | SUBSTITUTED PYRROLO [2, 3-b] PYRIDINE AND PYRAZOLO [3, 4-b] PYRIDINE DERIVATIVES AS PROTEIN KINASE INHIBITORS |
| WO2021093839A1 (en) | 2019-11-13 | 2021-05-20 | 南京明德新药研发有限公司 | Pyrrolopyrimidine compound as btk inhibitor and use thereof |
| WO2023110970A1 (en) * | 2021-12-14 | 2023-06-22 | Netherlands Translational Research Center Holding B.V | Macrocyclic btk inhibitors |
| WO2023110936A1 (en) * | 2021-12-14 | 2023-06-22 | Netherlands Translational Research Center Holding B.V | Reversible macrocyclic kinase inhibitors |
-
2024
- 2024-05-31 WO PCT/EP2024/065039 patent/WO2024246287A1/en active Pending
Patent Citations (22)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2013010380A1 (en) | 2011-07-19 | 2013-01-24 | Merck Sharp & Dohme Corp. | Btk inhibitors |
| WO2013067274A1 (en) | 2011-11-03 | 2013-05-10 | Genentech, Inc. | Heteroaryl pyridone and aza-pyridone compounds as inhibitors of btk activity |
| WO2013185084A1 (en) | 2012-06-08 | 2013-12-12 | Biogen Idec Ma Inc. | Pyrimidinyl tyrosine kinase inhibitors |
| WO2014018567A1 (en) * | 2012-07-24 | 2014-01-30 | Pharmacyclics, Inc. | Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk) |
| WO2016109222A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Tertiary alcohol imidazopyrazine btk inhibitors |
| WO2016106626A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Imidazopyrazine analogs with 3-tertiary carbon substitutions as btk inhibitors |
| WO2016106628A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Btk inhibitors |
| WO2016106624A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Tertiary alcohol imidazopyrazine btk inhibitors |
| WO2016106625A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Btk inhibitors |
| WO2016106623A1 (en) | 2014-12-31 | 2016-07-07 | Merck Sharp & Dohme Corp. | Benzamide imidazopyrazine btk inhibitors |
| WO2016161571A1 (en) | 2015-04-08 | 2016-10-13 | Merck Sharp & Dohme Corp. | Indazole and azaindazole btk inhibitors |
| WO2016161570A1 (en) | 2015-04-08 | 2016-10-13 | Merck Sharp & Dohme Corp. | Azacarbazole btk inhibitors |
| WO2017046604A1 (en) | 2015-09-16 | 2017-03-23 | Redx Pharma Plc | Pyrazolopyrimidine derivatives as btk inhibitors for the treatment of cancer |
| WO2017103611A1 (en) | 2015-12-16 | 2017-06-22 | Redx Pharma Plc | Compounds useful as kinase inhibitors |
| WO2017111787A1 (en) | 2015-12-23 | 2017-06-29 | Arqule, Inc. | Tetrahydropyranyl amino-pyrrolopyrimidinone and methods of use thereof |
| WO2018097234A1 (en) | 2016-11-25 | 2018-05-31 | カルナバイオサイエンス株式会社 | Novel oxoisoquinoline derivative |
| WO2020015735A1 (en) | 2018-07-20 | 2020-01-23 | 正大天晴药业集团股份有限公司 | Bruton tyrosine kinase inhibitors |
| WO2020043638A1 (en) | 2018-08-28 | 2020-03-05 | Merck Patent Gmbh | Fused imidazopyridines as reversible inhibitors of bruton's tyrosine kinase (btk) |
| WO2020239124A1 (en) | 2019-05-31 | 2020-12-03 | Fochon Pharmaceuticals, Ltd. | SUBSTITUTED PYRROLO [2, 3-b] PYRIDINE AND PYRAZOLO [3, 4-b] PYRIDINE DERIVATIVES AS PROTEIN KINASE INHIBITORS |
| WO2021093839A1 (en) | 2019-11-13 | 2021-05-20 | 南京明德新药研发有限公司 | Pyrrolopyrimidine compound as btk inhibitor and use thereof |
| WO2023110970A1 (en) * | 2021-12-14 | 2023-06-22 | Netherlands Translational Research Center Holding B.V | Macrocyclic btk inhibitors |
| WO2023110936A1 (en) * | 2021-12-14 | 2023-06-22 | Netherlands Translational Research Center Holding B.V | Reversible macrocyclic kinase inhibitors |
Non-Patent Citations (37)
| Title |
|---|
| "Remington's Pharmaceutical Sciences", 2000, MEADE PUBLISHING CO. |
| AL-LAZIKANI BBANERJI UWORKMAN P: "Combinatorial drug therapy for cancer in the post-genomic era", NATURE BIOTECH, vol. 30, 2012, pages 679 - 682 |
| ATTWOOD M.M. ET AL.: "Trends in kinase drug discovery targets, indications and inhibitor design", NAT REV DRUG DISCOV, vol. 20, 2021, pages 839 - 861, XP037602806, DOI: 10.1038/s41573-021-00252-y |
| BODOR ET AL., BRITISH JOURNAL OF HAEMATOLOGY, 2021 |
| BODOR, C., KOTMAYER, L., LÁSZLÓ, T., TAKACS, F., BARNA, G., KISS, R., SEBESTY6N, E.,NAGY, T., HEGYI, L., MIKALA, G., FEKETE, S. P.: "Screening and monitoring of the BTK C481 S mutation in a real-world cohort of patients with relapsed/refractory chronic lymphocytic leukaemia during ibrutinib therapy", JOURNAL OF HAEMATOLOGY, vol. 194, no. 2, 2021, pages 355 - 364 |
| BORISY AAELLIOTT PJHURST NWLEE MSLEHAR JPRICE ER ET AL.: "Systematic discovery of multicomponent therapeutics", PROC NATL ACAD SCI USA, vol. 100, 2003, pages 7977 - 7982, XP009061663, DOI: 10.1073/pnas.1337088100 |
| CHOU, T-C: "Drug combination studies and their synergy quantification using the Chou-Talalay method", CANCER RES, vol. 70, 2010, pages 440 - 446, XP055169871, DOI: 10.1158/0008-5472.CAN-09-1947 |
| COKOL M, CHUA HN, TASAN M, MUTLU B, WEINSTEIN ZB, SUZUKI Y: "Systematic exploration of synergistic drug pairs", MOLECULAR SYSTEMS BIOLOGY, vol. 7, 2011, pages 544 |
| CRAWFORD, J., JOHNSON, A. R., MISNER, D., BELMONT, L. D., CASTANEDO, G., CHOY, R.,CORAGGIO, M., DONG, L., EIGENBROT, C., ERICKSON,: "Discovery of GDC-0853: A Potent, Selective, and Noncovalent Bruton's Tyrosine Kinase Inhibitor in Early Clinical Development", CHEMISTRY, vol. 61, no. 6, 2018, pages 2227 - 2245, XP055685494, DOI: 10.1021/acs.jmedchem.7b01712 |
| GARNETT MJEDELMAN EJHEIDORN SJGREENMAN CDDASTUR ALAU KW ET AL.: "Systematic identification of genomic markers of drug sensitivity in cancer cells", NATURE, vol. 483, 2012, pages 570 - 575, XP055186003, DOI: 10.1038/nature11005 |
| GU DANLING ET AL: "Targeting Bruton tyrosine kinase using non-covalent inhibitors in B cell malignancies", vol. 14, no. 1, 6 March 2021 (2021-03-06), pages 40, XP055861303, Retrieved from the Internet <URL:https://jhoonline.biomedcentral.com/track/pdf/10.1186/s13045-021-01049-7.pdf> DOI: 10.1186/s13045-021-01049-7 * |
| HOPKINS, B., BAME, E., BAJRAMI, B., BLACK, C. A., BOHNERT, T., BOISELLE, C., BURDETTE, D.,BURNS, J. C., DELVA, L., DONALDSON, D., : "Discovery and Preclinical Characterization of BIIB091, a Reversible, Selective BTK Inhibitor for the Treatment of Multiple Sclerosis", JOURNAL OF MEDICINAL CHEMISTRY, vol. 65, no. 2, 2021, pages 1206 - 1224, XP093034944, DOI: 10.1021/acs.jmedchem.1c00926 |
| KAWAHATA, W., ASAMI, T., KIYOI, T., IRIE, T., KASHIMOTO, S., FURUICHI, H., & SAWA, M.: "Discovery of AS-1763: A Potent, Selective, Noncovalent, and Orally Available Inhibitor of Bruton's Tyrosine Kinase", JOURNAL OF MEDICINAL CHEMISTRY, vol. 64, no. 19, 2021, pages 14129 - 14141, XP055930428, DOI: 10.1021/acs.jmedchem.1c01279 |
| KAWAHATA, W.ASAMI, T.KIYOI, T.IRIE, T.TANIGUCHI, H.ASAMITSU, Y.INOUE, T.MIYAKE, T.SAWA, M.: "Design and Synthesis of Novel Amino-triazine Analogues as Selective Bruton's Tyrosine Kinase Inhibitors for Treatment of Rheumatoid Arthritis", JOURNAL OF MEDICINAL CHEMISTRY, vol. 61, no. 19, 2018, pages 8917 - 8933, XP093120071, DOI: 10.1021/acs.jmedchem.8b01147 |
| KOKABEE, L.WANG, X.SEVINSKY, C. J.WANG, W.CHEU, L.CHITTUR, S. V.KARIMIPOOR, M.TENNISWOOD, M.CONKLIN, D. S.: "Bruton's tyrosine kinase is a potential therapeutic target in prostate cancer", CANCER BIOLOGY & THERAPY, vol. 16, no. 11, 2015, pages 1604 - 1615 |
| LI FZHAO CWANG L: "Molecular-targeted agents combination therapy for cancer: developments and potentials", INT J CANCER, vol. 134, 2014, pages 1257 - 1269, XP055517584, DOI: 10.1002/ijc.28261 |
| LIU, Q.YIN, X.LANGUINO, L. R.ALTIERI, D. C.: "Evaluation of Drug Combination Effect Using a Bliss Independence Dose-Response Surface Model", STATISTICS IN BIOPHARMACEUTICAL RESEARCH, vol. 10, no. 2, 2018, pages 112 - 122, XP093053632, DOI: 10.1080/19466315.2018.1437071 |
| MATHEWS GRINER LAGUHA RSHINN PYOUNG RMKELLER JMLIU D ET AL.: "High throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells", PROC NATL ACAD SCI USA, vol. 111, 2014, pages 2349 - 2354, XP055218384, DOI: 10.1073/pnas.1311846111 |
| NAKHODA, S., VISTAROP, A., & WANG, Y. L.: "Resistance to Bruton tyrosine kinase inhibition in chronic lymphocytic leukaemia and non-Hodgkin lymphoma", OF HAEMATOLOGY, vol. 200, no. 2, 2022, pages 137 - 149 |
| R. BAROUCH-BENTOV AND K. SAUER: "Mechanisms of drug resistance in kinases", EXP. OPIN. INVEST. DRUGS, vol. 20, 2011, pages 153 - 208 |
| REIFF, S. D.MANTEL, R.SMITH, L.GREENE, J. E.MUHOWSKI, E. M.FABIAN, C. A.GOETTL, V. M.TRAN, M. T.HARRINGTON, B. K.ROGERS, K. A.: "The BTK Inhibitor ARQ 531 Targets Ibrutinib-Resistant CLL and Richter Transformation", CANCER DISCOVERY, vol. 8, no. 10, 2018, pages 1300 - 1315, XP055939114, DOI: 10.1158/2159-8290.CD-17-1409 |
| REIFF, S. D.MUHOWSKI, E. M.GUINN, D.LEHMAN, A.FABIAN, C. A.CHENEY, C.MANTEL, R.SMITH, L.JOHNSON, A. J.YOUNG, W. B.: "Noncovalent inhibition of C481S Bruton tyrosine kinase by GDC-0853: a new treatment strategy for ibrutinib-resistant CLL", BLOOD, vol. 132, no. 10, 2018, pages 1039 - 1049 |
| STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH |
| STRAETEMANS RO'BRIEN TWOUTERS LVAN DUN JJANICOT M ET AL.: "Design and analysis of drug combination experiments", BIOMETRICAL J, vol. 47, 2005, pages 299 - 308, XP071616797, DOI: 10.1002/bimj.200410124 |
| T.W. GREENEP.G.M. WUTTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS |
| THIEME, E.LIU, T.BRUSS, N.ROLEDER, C.LAM, V.WANG, X.NECHIPORUK, T.SHOUSE, G.DANILOVA, O. V.BOTTOMLY, D.: "Dual BTK/SYK inhibition with CG-806 (luxeptinib) disrupts B-cell receptor and Bcl-2 signaling networks in mantle cell lymphoma", CELL DEATH AND DISEASE, vol. 13, 2022, pages 3 |
| THOMPSON PHILIP A ET AL: "Pirtobrutinib: a new hope for patients with BTK inhibitor-refractory lymphoproliferative disorders", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 141, no. 26, 10 May 2023 (2023-05-10), pages 3137 - 3142, XP087346928, ISSN: 0006-4971, [retrieved on 20230510], DOI: 10.1182/BLOOD.2023020240 * |
| WANG ET AL., N. ENGL. J. MED., vol. 386, 2022, pages 735 - 43 |
| WANG, E. W., MI, X., THOMPSON, M. C., MONTOYA, S., NOTTI, R. Q., AFAGHANI, J.,DURHAM, B. H., PENSON, A. V., WITKOWSKI, M. T., LU, : "Mechanisms of Resistance to Noncovalent Bruton's Tyrosine Kinase Inhibitors", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 386, no. 8, 2022, pages 735 - 743, XP093103917, DOI: 10.1056/NEJMoa2114110 |
| WANG, X., KOKABEE, L., KOKABEE, M., & CONKLIN, D. S.: "Bruton's Tyrosine Kinase and Its Isoforms in Cancer", FRONTIERS IN CELL AND DEVELOPMENTAL BIOLOGY, 2021, pages 9 |
| WANG, X.WONG, J. W. H.SEVINSKY, C. J.KOKABEE, L.KHAN, F. M.SUN, Y.CONKLIN, D. S.: "Bruton's Tyrosine Kinase Inhibitors Prevent Therapeutic Escape in Breast Cancer Cells", MOLECULAR CANCER THERAPEUTICS, vol. 15, no. 9, 2016, pages 2198 - 2208, XP055345697, DOI: 10.1158/1535-7163.MCT-15-0813 |
| WATTERSON, S. H.DE LUCCA, G. V.SHI, Q.LANGEVINE, C. M.LIU, Q.BATT, D. G.BERTRAND, M. B.GONG, H.DAI, J.YIP, S.: "Discovery of 6-Fluoro-5-(R)-(3-(S)-(8-fluoro-1-methyl-2,4-dioxo-1,2-dihydroquinazolin-3(4H)-yl)-2-methylphenyl)-2-(S)-(2-hyd roxypropan-2-yl)-2, 3,4, 9-tetrahyd ro-1 H-carbazo le-8-carboxamide (BMS-986142", JOURNAL OF MEDICINAL CHEMISTRY, vol. 59, no. 19, 2016, pages 9173 - 9200 |
| WEI, L.SU, Y.LIN, C. M.CHAO, T. Y.HUANG, S.HUYNH, T. D.JAN, H. J.WHANG-PENG, J.CHIOU, J. F.WU, A. T.: "Preclinical investigation of ibrutinib, a Bruton's kinase tyrosine (Btk) inhibitor, in suppressing glioma tumorigenesis and stem cell phenotypes", ONCOTARGET, vol. 7, no. 43, 2016, pages 69961 - 69975, XP055649279, DOI: 10.18632/oncotarget.11572 |
| WOYACH, J. A.RUPPERT, A. S.GUINN, D.LEHMAN, A.BLACHLY, J. S.LOZANSKI, A.HEEREMA, N. A.ZHAO, W.COLEMAN, J. T.JONES, D. B.: "BTKC481 S-Mediated Resistance to Ibrutinib in Chronic Lymphocytic Leukemia", JOURNAL OF CLINICAL ONCOLOGY, vol. 35, no. 13, 2017, pages 1437 - 1443 |
| WOYACH, J. CLIN. ONCOL., vol. 35, 2017, pages 1437 - 1443 |
| YAP TAOMLIN ADE BONO JS: "Development of therapeutic combinations targeting major cancer signaling pathways", J CLINICAL INVEST, vol. 20, 2013, pages 1592 - 1605, XP009184619, DOI: 10.1200/JCO.2011.37.6418 |
| ZHAO L, WIENTJES MG, AU JLS: "Evaluation of combination chemotherapy:integration of nonlinear regression, curve shift, isobologram and combination index analyses", CLIN CANC RES, vol. 10, 2004, pages 7994 - 8004 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7337133B2 (en) | Pyrazolo[3,4-b]pyridine compounds as inhibitors of TAM and MET kinases | |
| US11452725B2 (en) | Chiral diaryl macrocycles and uses thereof | |
| TWI837478B (en) | Compounds for the treatment of braf-associated diseases and disorders | |
| DK2663564T3 (en) | IMIDAZO [4,5-C] QUINOLIN-2-ON COMPOUND AND ITS USE AS PI3-KINASE / MTOR DUAL INHIBITOR | |
| TW202136252A (en) | Compounds and uses thereof | |
| EP4247381A1 (en) | Compounds and uses thereof | |
| BR112017019790B1 (en) | USE OF A BRUTON TYROSINE KINASE (BTK) INHIBITOR | |
| EP4132529A1 (en) | Compounds and uses thereof | |
| CN106922146A (en) | Pyrazole carboxamide compounds for the treatment of diseases mediated by Bruton's tyrosine kinase (BTK) | |
| EP4448523A1 (en) | Macrocyclic btk inhibitors | |
| KR20250024923A (en) | Compounds and their uses | |
| KR20250020451A (en) | Benzoylparazine pyrazine and its uses | |
| WO2024246287A1 (en) | Medical use of a macrocyclic reversible btk inhibitor | |
| WO2024245578A1 (en) | Therapeutic combinations of an irreversible btk inhibitor and a macrocyclic reversible btk inhibitor | |
| RU2814662C1 (en) | 4-oxo-3,4-dihydroquinazolinone compounds for treatment of braf-associated diseases and disorders | |
| WO2025222187A1 (en) | Compounds and uses thereof | |
| WO2025222132A1 (en) | Compounds and uses thereof | |
| AU2023270000A1 (en) | Pyrazine derivatives and uses thereof | |
| WO2025222125A1 (en) | Compounds and uses thereof | |
| HK40030138A (en) | Pyrazole carboxamide compounds for use in the treament of disorders mediated by bruton's tyrosine kinase (btk) | |
| HK40055909A (en) | Pyrazolo[3,4-b]pyridine compounds as inhibitors of tam and met kinases | |
| BR112022024597B1 (en) | 4-OXO-3,4-DIHYDROQUINAZOLINONE COMPOUNDS FOR THE TREATMENT OF BRAF-ASSOCIATED DISEASES AND DISORDERS, COMPOSITIONS AND USES THEREOF | |
| HK40053753A (en) | Quinoline compounds as inhibitors of tam and met kinases | |
| HK1235057B (en) | Pyrazole carboxamide compounds for use in the treament of disorders mediated by bruton's tyrosine kinase (btk) | |
| HK1222858A1 (en) | Heteroaryl pyridone and aza-pyridone compounds with electrophilic functionality |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24731495 Country of ref document: EP Kind code of ref document: A1 |