WO2024108028A1 - Methods of use of deuterium-enriched compounds - Google Patents

Methods of use of deuterium-enriched compounds Download PDF

Info

Publication number
WO2024108028A1
WO2024108028A1 PCT/US2023/080129 US2023080129W WO2024108028A1 WO 2024108028 A1 WO2024108028 A1 WO 2024108028A1 US 2023080129 W US2023080129 W US 2023080129W WO 2024108028 A1 WO2024108028 A1 WO 2024108028A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
compound
formula
effective amount
pharmaceutically acceptable
Prior art date
Application number
PCT/US2023/080129
Other languages
French (fr)
Inventor
Aundrietta DUNCAN
Stephen K. Horrigan
Daniela SANTIESTEBAN
Original Assignee
Salarius Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Salarius Pharmaceuticals, Inc. filed Critical Salarius Pharmaceuticals, Inc.
Publication of WO2024108028A1 publication Critical patent/WO2024108028A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/63Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
    • A61K31/635Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide having a heterocyclic ring, e.g. sulfadiazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • Lymphomas including diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL), are one of the most common classes of cancer, accounting for approximately 4% of all cancers.
  • a number of drug compounds have been studied for the treatment of lymphomas (e.g., avadomide or lenalidomide).
  • Many of the currently approved drugs for treating lymphomas do not provide the required efficacy when delivered as a monotherapy.
  • lenalidomide and many of the drug candidates in development for lymphomas are commonly delivered as racemic mixtures, rather than as a pure enantiomeric form, which may result in lower efficacy and potentially contribute to adverse effects.
  • kits for treating a lymphoma in a subject in need thereof comprising administering to the subject a compound of formula (I) or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax.
  • the method comprises administering an effective amount of a combination of the compound of formula (I) and the second therapeutic agent.
  • the lymphoma is follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL).
  • FL follicular lymphoma
  • DLBCL diffuse large B- cell lymphoma
  • the lymphoma is FL.
  • the lymphoma is DLBCL.
  • the second therapeutic agent is tazemetostat. In certain embodiments, the second therapeutic agent is venetoclax. In certain embodiments, the second therapeutic agent is rituximab.
  • the method reduces tumor growth or volume in the subject. In certain embodiments, the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of the second therapeutic agent.
  • a method of treating FL in a subject in need thereof comprises administering to the subject an effective amount of a combination of a compound of formula (I) or a pharmaceutically acceptable salt thereof, and tazemetostat.
  • the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat.
  • a method of treating FL in a subject in need thereof comprises administering to the subject an effective amount of a combination of a compound of formula or a pharmaceutically acceptable salt thereof, and venetoclax.
  • the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax.
  • a method of treating DLBCL in a subject in need thereof comprises administering to the subject an effective amount of a combination of a compound of formula (I)
  • the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab.
  • the compound of formula (I), or pharmaceutically acceptable salt thereof is administered orally.
  • the compound of formula (I) is administered in its free base form.
  • FIG. 1 is an overlay of tumor volume (mm 3 ) data over time for (i) vehicle; (ii) SP-3164 (15 mg/kg BID); (iii) lenalidomide (30 mg/kg BID); (iv) rituximab (10 mg/kg BIW); (v) SP- 3164 (15 mg/kg BID) + rituximab (10 mg/kg BIW); and (vi) lenalidomide (30 mg/kg BID) + rituximab (10 mg/kg BIW) administered to WSU-DLCL2 xenograft mice in a mouse diffuse large B-cell lymphoma (DLBCL) model (**** p ⁇ 0.0001), as described in Example 3.
  • DLBCL mouse diffuse large B-cell lymphoma
  • FIG. 2 is an overlay of tumor volume (mm 3 ) data over time for (i) vehicle; (ii) SP-3164 (7.5 mg/kg BID); (iii) tazemetostat (300 mg/kg BID); and (iv) SP-3164 (7.5 mg/kg BID) + tazemetostat (300 mg/kg BID) administered to DOHH2 xenograft mice in a mouse follicular lymphoma (FL) model (*** p ⁇ 0.001), as described in Example 4.
  • FL mouse follicular lymphoma
  • FIG. 3 is an overlay of tumor volume (mm 3 ) data over time for (i) vehicle; (ii) SP-3164 (7.5 mg/kg BID); (iii) venetoclax (50 mg/kg QD 5 on/2 off); and SP-3164 (7.5 mg/kg BID) + venetoclax (50 mg/kg QD 5 on/2 off) administered to DOHH2 xenograft mice in a mouse FL model (*** p ⁇ 0.001), as described in Example 4.
  • the present disclosure provides methods of treating a lymphoma (e.g., follicular lymphoma (FL) or diffuse large B-cell lymphoma (DLBCL)) in a subject in need thereof.
  • the methods generally comprise administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful for the treatment of lymphoma (e.g., rituximab, tazemetostat, and venetoclax).
  • values are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual subcombination of the members of such groups and ranges.
  • an integer in the range of 0 to 40 is specifically intended to individually disclose 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, and 40
  • an integer in the range of 1 to 20 is specifically intended to individually disclose 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20.
  • the terms “subject” and “patient” refer to organisms to be treated by the methods and/or compositions described herein. Such organisms are preferably mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and more preferably humans.
  • a “compound” refers to the compound itself and its pharmaceutically acceptable salts unless otherwise understood from the context of the description or expressly limited to one particular form of the compound, e.g., the compound itself, or a pharmaceutically acceptable salt thereof.
  • the compounds herein described may have asymmetric centers, geometric centers (e.g., double bond), or both. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated.
  • Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms, by synthesis from optically active starting materials, or through use of chiral auxiliaries.
  • cis and trans geometric isomers of the compounds of the present invention may also exist and may be isolated as a mixture of isomers or as separated isomeric forms. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. All tautomers of shown or described compounds are also considered to be part of the present invention.
  • pharmaceutically acceptable refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient. Accordingly, pharmaceutically acceptable carriers are those that are compatible with the other ingredients in the formulation and are biologically acceptable. In certain embodiments, supplementary active ingredients can also be incorporated into the pharmaceutical compositions.
  • pharmaceutically acceptable excipient refers to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present invention without causing a significant adverse toxicological effect on the patient.
  • Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, a phosphate buffered saline solution, emulsions (e.g., such as an oil/water or water/oil emulsions), lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer’s solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxypropylmethylcellulose, polyvinyl pyrrolidine, and colors, and the like.
  • emulsions e.g., such as an oil/water or water/oil emulsions
  • lactated Ringer lactated Ringer’s
  • sucrose normal glucose
  • binders fillers
  • disintegrants e.g., such as an oil/water or water/oil emul
  • Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
  • the term “pharmaceutically acceptable salt” refers to any pharmaceutically acceptable salt (e.g., acid or base) of a compound of the present invention (e.g., a compound of formula (I)) which, upon administration to a subject, is capable of providing a compound of this invention or an active metabolite or residue thereof.
  • salts of the compounds of the present invention may be derived from inorganic or organic acids and bases.
  • acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like.
  • Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW4+, wherein W is Cl -4 alkyl, and the like.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • W is Cl -4 alkyl
  • salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate,
  • the term “effective amount” refers to the amount of a compound provided herein (e.g., compound of formula (I), or a pharmaceutically acceptable salt thereof, or a combination of compounds) sufficient to effect beneficial or desired results.
  • An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • administering a “combination” of compounds refers to two or more compounds being administered to the same patient as part of the same treatment.
  • the compounds in the combination can be administered as a single dosage form or as separate dosage forms.
  • the compounds in the combination can be administered simultaneously or sequentially. Sequential administration of the compounds in the combination can be on the same day or separate days.
  • the terms “treat,” “treating,” and “treatment” include any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
  • compositions and kits are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions and kits of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.
  • Prevent refers to the prevention of the onset, recurrence or spread of a disease or disorder, or of one or more symptoms thereof.
  • Deuterium (D or 2 H) is a stable, non-radioactive isotope of hydrogen and has an atomic weight of 2.014. Hydrogen naturally occurs as a mixture of the isotopes 1 H (hydrogen or protium), D ( 2 H or deuterium), and T ( 3 H or tritium). The natural abundance of deuterium is 0.015%.
  • the H atom actually represents a mixture of H and D, with about 0.015% being D.
  • compounds with a level of deuterium that has been enriched to be greater than its natural abundance of 0.015% should be considered unnatural and, as a result, novel over their nonenriched counterparts.
  • the present invention relates to a deuterium-enriched compound or compounds whose enrichment is greater than naturally occurring deuterated molecules.
  • this D represents a mixture of hydrogen and deuterium where the amount of deuterium is about 100% (i.e., the abundance of deuterium is from 80% to 100%). In certain aspects, the abundance of deuterium is from 97% to 100%.
  • Compound refers to a quantity of molecules that is sufficient to be weighed, tested for its structural identity, and to have a demonstrable use (e.g., a quantity that can be shown to be active in an assay, an in vitro test, or in vivo test, or a quantity that can be administered to a patient and provide a therapeutic benefit).
  • “lenalidomide” refers to a compound having the formula (II): or pharmaceutically acceptable salts thereof. Such salts may include, but are not limited to, lenalidomide benzenesulfonate and lenalidomide p-toluenesulfonate. Lenalidomide may be referred to by other names, for example, its IUPAC name: 3-(4-amino-l-oxoisoindolin-2- yl)piperidine-2, 6-dione, its CAS number: 191732-72-6, Revimid, or its common brand name: Revlimid®.
  • “rituximab” refers to a chimeric monoclonal antibody targeted against CD20. Rituximab may be referred to by other names, for example, its CAS number: 174722-31-
  • tazemetostat refers to a compound of formula (III): or pharmaceutically acceptable salts thereof. Such salts may include, but are not limited to, tazemetostat hydrobromide.
  • Tazemetostat may be referred to by other names, for example, its IUPAC name: A-[(4,6-dimethyl-2-oxo-177-pyridin-3-yl)methyl]-3-[ethyl(oxan-4-yl)amino]-2- methyl-5-[4-(morpholin-4-ylmethyl)phenyl]benzamide, its CAS number: 1403254-99-8, EPZ- 6438, or its common brand name: Tazverik®.
  • Venetoclax refers to a compound of formula (IV): or pharmaceutically acceptable salts thereof. Such salts may include, but are not limited to, venetoclax docusate. Venetoclax may be referred to by other names, for example, its IUPAC name: 4-[4-[[2-(4-chlorophenyl)-4,4-dimethylcyclohexen-l-yl]methyl]piperazin-l-yl]-7V-[3- nitro-4-(oxan-4-ylmethylamino)phenyl]sulfonyl-2-(177-pyrrolo[2,3-b]pyridin-5- yloxy)benzamide, its CAS number: 1257044-40-8, or its common brand names: Venclexta® and Venclyxto®.
  • IUPAC name 4-[4-[[2-(4-chlorophenyl)-4,4-dimethylcyclohexen-l-yl]methyl]piperazin-l
  • the compound of formula (I), as depicted below, is a deuterium-enriched compound, and is also known as (N)-3-(5-amino-2-methyl-4-oxoquinazolin-3(4//)-yl)-(3- 2 H)-piperidine-2,6- dione: or a pharmaceutically acceptable salt thereof.
  • the compound of formula (I) may also be referred to as “SP-3164” or “deuterated S- avadomide” throughout the present disclosure.
  • SP-3164 or “deuterated S- avadomide” throughout the present disclosure.
  • a method of chemically synthesizing the compound of formula (I) is described in U.S. Patent No. 9,540,340, which is incorporated by reference in its entirety herein.
  • the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 85% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 90% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 95%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 90%.
  • the compound of formula (I) may also be characterized according to its stereochemical purity.
  • the stereochemical purity of a compound containing one stereocenter can be characterized as enantiomeric excess (ee).
  • the compound of formula (I) has an enantiomeric excess of at least 5%.
  • the compound for formula (I) has an enantiomeric excess of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%.
  • stereoisomeric purity examples include an enantiomeric excess of at least about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about 70%,
  • the compound of formula (I) has a stereomeric purity (enantiomeric excess) of at least 5%.
  • the compound of formula (I) has a stereomeric purity (enantiomeric excess) of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%.
  • stereoisomeric purity (enantiomeric excess) of the compound of formula (I) include at least about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 65%
  • the compound of formula (I) has an optical purity of at least 85% enantiomeric excess, 90% enantiomeric excess, 95% enantiomeric excess, or 98% enantiomeric excess.
  • a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent e.g., rituximab, tazemetostat, or venetoclax
  • a lymphoma e.g., follicular lymphoma (FL) or diffuse large B-cell lymphoma (DLBCL)
  • FL follicular lymphoma
  • DLBCL diffuse large B-cell lymphoma
  • a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent e.g., rituximab, tazemetostat, or venetoclax
  • a lymphoma e.g., FL or DLBCL
  • provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab for the treatment of DLBCL in a subject in need thereof.
  • provided herein is a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab for the treatment of DLBCL in a subject in need thereof.
  • provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat for the treatment of FL in a subject in need thereof.
  • provided herein is a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat for the treatment of FL in a subject in need thereof.
  • provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax for the treatment of FL in a subject in need thereof.
  • compositions generally comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
  • a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) a second therapeutic agent (e.g., rituximab, tazemetostat, or venetoclax) for the treatment of a lymphoma (e.g., follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL)) in a subject in need thereof.
  • a lymphoma e.g., follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL)
  • a method of administering a combination of (i) a pharmaceutical composition comprising an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) an effective amount of a second therapeutic agent (e.g., rituximab, tazemetostat, or venetoclax) for the treatment of a lymphoma (e.g., FL or DLBCL) in a subject in need thereof.
  • a second therapeutic agent e.g., rituximab, tazemetostat, or venetoclax
  • a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) rituximab for the treatment of DLBCL in a subject in need thereof.
  • a method of administering a combination of (i) a pharmaceutical composition comprising an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) an effective amount of rituximab for the treatment of DLBCL in a subject in need thereof.
  • provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) tazemetostat for the treatment of FL in a subject in need thereof.
  • provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) venetoclax for the treatment of FL in a subject in need thereof.
  • compositions described herein can be administered by a variety of routes including, but not limited to, oral (enteral) administration, parenteral (by injection) administration, rectal administration, transdermal administration, intradermal administration, intrathecal administration, subcutaneous (SC) administration, intravenous (IV) administration, intramuscular (IM) administration, and intranasal administration.
  • oral (enteral) administration parenteral (by injection) administration
  • rectal administration transdermal administration
  • intradermal administration intrathecal administration
  • SC subcutaneous
  • IV intravenous
  • IM intramuscular
  • intranasal administration intranasal administration.
  • the pharmaceutical compositions described herein are administered orally.
  • the pharmaceutical compositions described herein may also be administered chronically (“chronic administration”).
  • Chronic administration refers to administration of a compound or pharmaceutical composition thereof over an extended period of time, e.g., for example, over 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may be continued indefinitely, for example, for the rest of the subject’s life.
  • the chronic administration is intended to provide a constant level of the compound in the blood, e.g., within the therapeutic window over the extended period of time.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with ordinary experimentation. General considerations in the formulation and/or manufacture of pharmaceutical compositions can be found, for example, in Remington : The Science and Practice of Pharmacy 21 st ed. , Lippincott Williams & Wilkins, 2005.
  • provided herein are methods of treating a lymphoma in a subject in need thereof.
  • a method of treating a lymphoma in a subject in need thereof comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful for the treatment of lymphomas.
  • a method of treating a lymphoma in a subject in need thereof comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful for the treatment of lymphomas.
  • the second therapeutic agent useful for the treatment of lymphoma is selected from the group consisting of rituximab, tazemetostat, and venetoclax.
  • a method of treating a lymphoma in a subject in need thereof comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax.
  • a method of treating a lymphoma in a subject in need thereof comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax.
  • the lymphoma is follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL).
  • the lymphoma is FL.
  • the lymphoma is DLBCL.
  • the second therapeutic agent is rituximab. In certain embodiments, the second therapeutic agent is tazemetostat. In certain embodiments, the second therapeutic agent is tazemetostat.
  • the method reduces tumor volume or the rate of tumor growth in the subject. In certain embodiments, the method reduces tumor volume in the subject. In certain embodiments, the method reduces the rate of tumor growth in the subject.
  • the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of the second therapeutic agent.
  • the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a 3) subject receiving only the effective amount of the second therapeutic agent.
  • the method reduces the tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of the second therapeutic agent.
  • the second therapeutic agent is administered orally. In certain embodiments, the second therapeutic agent is administered intravenously. In certain embodiments, the second therapeutic agent is administered by intravenous infusion.
  • a method of treating FL in a subject in need thereof comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat.
  • the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat.
  • the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat.
  • the method reduces tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat.
  • a method of treating FL in a subject in need thereof comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax.
  • the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax.
  • the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax.
  • the method reduces tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax.
  • a method of treating DLBCL in a subject in need thereof comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab.
  • the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab.
  • the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab.
  • the method reduces tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab.
  • the tazemetostat is administered orally.
  • the venetoclax is administered orally.
  • the rituximab is administered intravenously.
  • the rituximab is administered by intravenous infusion.
  • the compound of formula (I), or a pharmaceutically acceptable salt thereof is administered orally.
  • the compound of formula (I) is administered in its free base form. In certain embodiments, the compound of formula (I) is administered as a pharmaceutically acceptable salt. [00101] In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 85% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 90% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 95%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 90%.
  • a method of treating a lymphoma comprising administering to the subject a pharmaceutical composition described herein and a second therapeutic agent (e.g., rituximab, tazemetostat, and venetoclax) useful in the treatment of lymphoma.
  • a lymphoma e.g., FL or DLBCL
  • a second therapeutic agent e.g., rituximab, tazemetostat, and venetoclax
  • a method of treating a lymphoma comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and a second therapeutic agent (e.g., rituximab, tazemetostat, and venetoclax) useful in the treatment of lymphoma.
  • a lymphoma e.g., FL or DLBCL
  • a second therapeutic agent e.g., rituximab, tazemetostat, and venetoclax
  • provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject a pharmaceutical composition described herein and tazemetostat.
  • provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and tazemetostat.
  • provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject a pharmaceutical composition described herein and venetoclax.
  • provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and venetoclax.
  • a method of treating DLBCL in a subject in need thereof comprising administering to the subject a pharmaceutical composition described herein and rituximab.
  • a method of treating DLBCL in a subject in need thereof comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and rituximab.
  • Certain combinations described herein may work synergistically in the treatment of the diseases, disorders, and conditions described herein (e.g., a lymphoma) and the symptoms associated with such diseases or disorders.
  • TGI Tumor Growth Inhibition (1-Ti/Ci) x 100;
  • Ti and Ci are the mean tumor volumes of the treatment and control groups, respectively, on a given day.
  • T/C Ti/Ci x 100;
  • Ti and Ci are the mean tumor volumes of the treatment and control groups, respectively, on a given day.
  • Step 2 Synthesis of 2-Acetamido-6-nitrobenzoic acid [00113]
  • the starting material (24.7 g, 120 mmol) was dispersed in water (216 mL) and the resulting mixture was heated to reflux for 30 min. Upon cooling the reaction mixture, crystallization began. Next, the reaction mixture was placed in a refrigerator for 18 hours to complete crystallization. The crystalline material was isolated by filtration and dried under vacuum to give the title compound (24.3 g, 108 mmol, 90.5%) as a pale yellow, crystalline solid.
  • the starting acid (3.10 g, 13.8 mmol) was mixed with hydroxybenzotriazole (HOBt, 2.12 g of the hydrate, 13.8 mmol) and l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC, 2.54 g, 13.3 mmol), under a nitrogen atmosphere.
  • HOBt hydroxybenzotriazole
  • EDC l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride
  • N,N-dimethylformamide (DMF, 21.4 mL) was added and the mixture was stirred for 30 minutes at room temperature, rac- 3-Aminopiperidine-2, 6-dione hydrochloride (5.01 g, 30.4 mmol) was added, followed by N,N- diisopropylethylamine (DIEA, 9.63 mL, 55.3 mmol).
  • DIEA N,N- diisopropylethylamine
  • the reaction mixture was stirred at 20 °C, while monitoring by HPLC. After 24 hours, the reaction mixture showed approximately 40% conversion to the desired product containing some remaining starting acid, but no amine. Then, the reaction mixture was slowly poured into 200 mL water with vigorous stirring. After 20 minutes, a white precipitate began to form. The mixture was placed in the refrigerator for 18 hours.
  • reaction mixture was cooled to 20 °C, stirred rapidly, and quenched with deuterium oxide (100 mL, 5.55 moles). The reaction mixture was stirred for an additional 20 minutes and a white precipitate formed. Next, the reaction mixture was cooled in a refrigerator for 4 hours and filtered to provide the title compound (2.25 g, 7.1 mmol, 53%) as an off-white, crystalline solid.
  • mice with an NOD/SCID genetic profile (6-8 weeks estimated average age at inoculation; Beijing Vital River Laboratory Animal Technology Co., Ltd) were used in the study.
  • mice were housed in polysulfone individually ventilated cages (IVC) cages (335mm x 143mm x 130mm, up to 5 per cage), supplied with autoclaved crushed corncob bedding that was changed weekly. The temperature and humidity were maintained at 20 - 26 °C and 40 - 70% humidity. A light cycle of 12 hours light and 12 hours dark was used. Mice were fed standard rodent chow (irradiated) and autoclaved, filtered (reverse osmosis) softened water ad libitum. Each mouse was identified by a labeled ear tag.
  • IVC polysulfone individually ventilated cages
  • Cages were identified by label, which included project number, group number, number of animals, gender, strain, supplier, date of receipt, treatment, animal use protocol (AUP) number, and date of treatment initiation. Daily cage side and weekly clinical observations were made. The protocol was reviewed and approved by the IACUC. During the study, the care and use of animals was conducted in accordance with the regulations of the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC). [00126] The study was designed to compare treatment effects of lenalidomide, SP-3164, and rituximab monotherapies and to compare combination therapies of lenalidomide and rituximab and SP-3164 and rituximab in a WSU-DLCL2 DLBCL xenograft model. Lenalidomide and SP- 3164 were administered orally (PO) twice daily (BID with an interval of 5h between the two doses each day). Rituximab was given intravenously (IV) bi-weekly (BIW).
  • IV bi-week
  • Each mouse was inoculated subcutaneously in the right upper flank region with WSU- DLCL2 cells (lx 10 7 ) in 0.1 ml of PBS mixed with Matrigel (1 : 1) for tumor development.
  • Treatment was initiated on the day of randomization (Day 0) per study design.
  • Day 0 Day 0
  • the animals were checked daily for morbidity and mortality.
  • body weight gain/loss body weights were measured twice per week after randomization
  • eye/hair matting eye/hair matting and any other abnormalities.
  • Mortality and observed clinical signs were recorded for individual animals.
  • Dosing as well as tumor and body weight measurements were conducted in a laminar flow cabinet. The body weights and tumor volumes were recorded by using StudyDirectorTM (version 3.1.399.19).
  • animals were scheduled to be euthanized if they lost more than 20% of their body weight relative to their weight on the first day of treatment. Similarly, animals were to be euthanized if their tumor volume were to exceed 3000 mm 3 .
  • any animal exhibiting an ulcerated or necrotic tumor was immediately separated, singly housed, and monitored daily before euthanasia or until tumor regression was complete. If the tumor ulcerated, and the ulceration diameter was greater than 5 mm, or pus or necrosis was observed; or if the tumor burden, including metastasis, compromised an animal’s normal physiologic performances, (e.g., orientation, access to food or water), the animal was euthanized rapidly.
  • a mouse was euthanized rapidly if its general welfare was deemed unacceptable (e.g., emaciated (skeletal structure extremely prominent, little or no flesh cover, vertebrae distinctly segmented); persistent hypothermia; blood-stained or mucopurulent discharge from any orifice; labored breathing, particularly accompanied by nasal discharge or cyanosis; hind limb paralysis or weakness (cannot reach food and water); severe anemia (mainly indicated by pale feet or ear); severe infection; or incontinence or diarrhea over 48 hours or severe dehydration).
  • Mouse treatment was performed for 28 days. Following cessation of treatment, animals and tumor growth were monitored until a humane termination endpoint was reached.
  • TGI analysis where the calculation was based on absolute tumor volume and relative tumor volume (RTV) was applied to the final data interpretation.
  • T/C analysis on the absolute T/C ratio and relative T/C ratio (RTV) was applied to the final data interpretation.
  • Survival (Kaplan-Meier model with log-rank method) analysis was applied to the final data interpretation. To compare tumor volumes differences amongst groups unpaired, two-tailed Student’s ttests were performed and p values ⁇ 0.05 regarded as statistically significant.
  • mice were fed standard rodent chow (irradiated) and autoclaved filtered (reverse osmosis) softened water ad libitum. Each mouse was identified by a labeled ear tag. Cages were identified by label, which included the project number, group number, number of animals, gender, strain, supplier, date of receipt, treatment, AUP number, and date of treatment initiation. Cage sides were monitored daily in conjunction with weekly clinical observations.
  • DOHH-2 cells were maintained in vitro with RPMI1640 medium supplemented with 10% fetal bovine serum at 37 °C in an atmosphere of 5% CO2 in the air. Cells in exponential growth phase were harvested and quantitated by cell counter before tumor inoculation.
  • Each mouse was inoculated subcutaneously in the right upper flank region with DOHH-2 cells (5 x 10 6 ) in 0.1 ml of PBS mixed with Matrigel (1 :1) for tumor development.
  • FIG. 2 shows that SP-3164 (7.5 mg/kg BID PO) demonstrated significant tumor growth inhibition (TGI) compared to vehicle and out-performed tazemetostat monotherapy (300 mg/kg BID PO).
  • TGI tumor growth inhibition
  • FIG. 3 shows that SP-3164 (7.5 mg/kg BID PO) showed significant TGI compared to vehicle.
  • the combination of SP-3164 and venetoclax demonstrated a synergistic TGI effect when compared to venetoclax or SP-3164 alone.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided herein are methods of treating a lymphoma in a subject in need thereof by administering to the subject a compound of formula (I) or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful in the treatment of a lymphoma.

Description

METHODS OF USE OF DEUTERI UM-ENRIC I ED COMPOUNDS
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of, and priority to, U.S. Provisional Application Serial No. 63/425,898, filed on November 16, 2022, the contents of which are incorporated by reference herein in their entirety.
BACKGROUND
[0002] Lymphomas, including diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL), are one of the most common classes of cancer, accounting for approximately 4% of all cancers. A number of drug compounds have been studied for the treatment of lymphomas (e.g., avadomide or lenalidomide). Many of the currently approved drugs for treating lymphomas do not provide the required efficacy when delivered as a monotherapy. Importantly, lenalidomide and many of the drug candidates in development for lymphomas are commonly delivered as racemic mixtures, rather than as a pure enantiomeric form, which may result in lower efficacy and potentially contribute to adverse effects. Further, administration of enantiopure compounds containing an unstable chiral center can lead to in vivo interconversion between enantiomeric forms resulting in the production of the undesired enantiomeric form, leading to the same unwanted outcomes as administering a racemic mixture. Therefore, there is an unmet need for the development of new therapeutic strategies for the treatment of lymphomas with single, stabilized enantiomers.
SUMMARY
[0003] In one aspect, provided herein are methods of treating a lymphoma in a subject in need thereof, the methods generally comprising administering to the subject a compound of formula (I)
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax.
[0004] In various embodiments, the method comprises administering an effective amount of a combination of the compound of formula (I) and the second therapeutic agent.
[0005] In certain embodiments, the lymphoma is follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL). In certain embodiments, the lymphoma is FL. In certain embodiments, the lymphoma is DLBCL.
[0006] In certain embodiments, the second therapeutic agent is tazemetostat. In certain embodiments, the second therapeutic agent is venetoclax. In certain embodiments, the second therapeutic agent is rituximab.
[0007] In certain embodiments, the method reduces tumor growth or volume in the subject. In certain embodiments, the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of the second therapeutic agent.
[0008] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, the method comprises administering to the subject an effective amount of a combination of a compound of formula (I)
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, and tazemetostat.
[0009] In certain embodiments, the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat.
[0010] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, the method comprises administering to the subject an effective amount of a combination of a compound of formula
Figure imgf000005_0002
or a pharmaceutically acceptable salt thereof, and venetoclax.
[0011] In certain embodiments, the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax.
[0012] In various embodiments, provided herein is a method of treating DLBCL in a subject in need thereof, the method comprises administering to the subject an effective amount of a combination of a compound of formula (I)
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof, and rituximab.
[0013] In certain embodiments, the method reduces tumor growth or volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab.
[0014] In certain embodiments, the compound of formula (I), or pharmaceutically acceptable salt thereof, is administered orally.
[0015] In certain embodiments, the compound of formula (I) is administered in its free base form.
BRIEF DESCRIPTION OF THE DRAWINGS
[0016] FIG. 1 is an overlay of tumor volume (mm3) data over time for (i) vehicle; (ii) SP-3164 (15 mg/kg BID); (iii) lenalidomide (30 mg/kg BID); (iv) rituximab (10 mg/kg BIW); (v) SP- 3164 (15 mg/kg BID) + rituximab (10 mg/kg BIW); and (vi) lenalidomide (30 mg/kg BID) + rituximab (10 mg/kg BIW) administered to WSU-DLCL2 xenograft mice in a mouse diffuse large B-cell lymphoma (DLBCL) model (**** p < 0.0001), as described in Example 3.
[0017] FIG. 2 is an overlay of tumor volume (mm3) data over time for (i) vehicle; (ii) SP-3164 (7.5 mg/kg BID); (iii) tazemetostat (300 mg/kg BID); and (iv) SP-3164 (7.5 mg/kg BID) + tazemetostat (300 mg/kg BID) administered to DOHH2 xenograft mice in a mouse follicular lymphoma (FL) model (*** p < 0.001), as described in Example 4.
[0018] FIG. 3 is an overlay of tumor volume (mm3) data over time for (i) vehicle; (ii) SP-3164 (7.5 mg/kg BID); (iii) venetoclax (50 mg/kg QD 5 on/2 off); and SP-3164 (7.5 mg/kg BID) + venetoclax (50 mg/kg QD 5 on/2 off) administered to DOHH2 xenograft mice in a mouse FL model (*** p < 0.001), as described in Example 4. DETAILED DESCRIPTION
[0019] As generally described herein, the present disclosure provides methods of treating a lymphoma (e.g., follicular lymphoma (FL) or diffuse large B-cell lymphoma (DLBCL)) in a subject in need thereof. The methods generally comprise administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful for the treatment of lymphoma (e.g., rituximab, tazemetostat, and venetoclax).
Definitions
[0020] To facilitate an understanding of the present invention, a number of terms and phrases are defined below. The examples provided in the definitions section as well as the remainder of this application are non-inclusive unless otherwise stated. They include but are not limited to the recited examples.
[0021] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The abbreviations used herein have their conventional meaning within the chemical and biological arts. The chemical structures and formulae set forth herein are constructed according to the standard rules of chemical valency known in the chemical arts.
[0022] The terms “a” and “an” as used herein mean “one or more” and include the plural unless the context is inappropriate.
[0023] In the application, where an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components, or the element or component can be selected from a group consisting of two or more of the recited elements or components.
[0024] Further, it should be understood that elements and/or features of a composition or a method described herein can be combined in a variety of ways without departing from the spirit and scope of the present invention, whether explicit or implicit herein. For example, where reference is made to a particular compound, that compound can be used in various embodiments of compositions of the present invention and/or in methods of the present invention, unless otherwise understood from the context. In other words, within this application, embodiments have been described and depicted in a way that enables a clear and concise application to be written and drawn, but it is intended and will be appreciated that embodiments may be variously combined or separated without parting from the present teachings and invention(s). For example, it will be appreciated that all features described and depicted herein can be applicable to all aspects of the invention(s) described and depicted herein.
[0025] It should be understood that the expression “at least one of’ includes individually each of the recited objects after the expression and the various combinations of two or more of the recited objects unless otherwise understood from the context and use. The expression “and/or” in connection with three or more recited objects should be understood to have the same meaning unless otherwise understood from the context.
[0026] The use of the term “include,” “includes,” “including,” “have,” “has,” “having,” “contain,” “contains,” or “containing,” including grammatical equivalents thereof, should be understood generally as open-ended and non-limiting, for example, not excluding additional unrecited elements or steps, unless otherwise specifically stated or understood from the context. [0027] Where the use of the term “about” is before a quantitative value, the present invention also includes the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term “about” refers to a ±10% variation from the nominal value unless otherwise indicated or inferred from the context.
[0028] At various places in the present specification, values are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual subcombination of the members of such groups and ranges. For example, an integer in the range of 0 to 40 is specifically intended to individually disclose 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, and 40, and an integer in the range of 1 to 20 is specifically intended to individually disclose 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20.
[0029] The use of any and all examples, or exemplary language herein, for example, “such as” or “including,” is intended merely to illustrate better the present invention and does not pose a limitation on the scope of the invention unless claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the present invention. [0030] As used herein, the terms “subject” and “patient” refer to organisms to be treated by the methods and/or compositions described herein. Such organisms are preferably mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and more preferably humans.
[0031] As used herein, a “compound” (including a specifically named compound, e.g., a compound of formula (I)) refers to the compound itself and its pharmaceutically acceptable salts unless otherwise understood from the context of the description or expressly limited to one particular form of the compound, e.g., the compound itself, or a pharmaceutically acceptable salt thereof.
[0032] The compounds herein described may have asymmetric centers, geometric centers (e.g., double bond), or both. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms, by synthesis from optically active starting materials, or through use of chiral auxiliaries. Geometric isomers of olefins, C=N double bonds, or other types of double bonds may be present in the compounds described herein, and all such stable isomers are included in the present invention. Specifically, cis and trans geometric isomers of the compounds of the present invention may also exist and may be isolated as a mixture of isomers or as separated isomeric forms. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. All tautomers of shown or described compounds are also considered to be part of the present invention.
[0033] As used herein, “pharmaceutically acceptable” refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient. Accordingly, pharmaceutically acceptable carriers are those that are compatible with the other ingredients in the formulation and are biologically acceptable. In certain embodiments, supplementary active ingredients can also be incorporated into the pharmaceutical compositions.
[0034] As used herein, “pharmaceutically acceptable excipient” refers to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present invention without causing a significant adverse toxicological effect on the patient. Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, a phosphate buffered saline solution, emulsions (e.g., such as an oil/water or water/oil emulsions), lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer’s solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxypropylmethylcellulose, polyvinyl pyrrolidine, and colors, and the like. Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention. For examples of excipients and carriers, see Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA (1975).
[0035] As used herein, the term “pharmaceutically acceptable salt” refers to any pharmaceutically acceptable salt (e.g., acid or base) of a compound of the present invention (e.g., a compound of formula (I)) which, upon administration to a subject, is capable of providing a compound of this invention or an active metabolite or residue thereof. As is known to those of skill in the art, “salts” of the compounds of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
[0036] Examples of bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW4+, wherein W is Cl -4 alkyl, and the like.
[0037] Examples of salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, undecanoate, and the like. Other examples of salts include anions of the compounds of the present invention compounded with a suitable cation such as Na+, NH4+, and NW4+ (wherein W is a Cl -4 alkyl group), and the like.
[0038] As used herein, the term “effective amount” refers to the amount of a compound provided herein (e.g., compound of formula (I), or a pharmaceutically acceptable salt thereof, or a combination of compounds) sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
[0039] As used herein, administering a “combination” of compounds refers to two or more compounds being administered to the same patient as part of the same treatment. The compounds in the combination can be administered as a single dosage form or as separate dosage forms. The compounds in the combination can be administered simultaneously or sequentially. Sequential administration of the compounds in the combination can be on the same day or separate days. [0040] As used herein, the terms “treat,” “treating,” and “treatment” include any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
[0041] Throughout the description, where compositions and kits are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions and kits of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.
[0042] " Prevent", "preventing" and "prevention" refer to the prevention of the onset, recurrence or spread of a disease or disorder, or of one or more symptoms thereof.
[0043] "Manage", "managing" and "management" refer to preventing or slowing the progression, spread or worsening of a disease or disorder, or of one or more symptoms thereof. In certain cases, the beneficial effects that a subject derives from a prophylactic or therapeutic agent do not result in a cure of the disease or disorder. [0044] Deuterium (D or 2H) is a stable, non-radioactive isotope of hydrogen and has an atomic weight of 2.014. Hydrogen naturally occurs as a mixture of the isotopes 1 H (hydrogen or protium), D (2H or deuterium), and T (3H or tritium). The natural abundance of deuterium is 0.015%. One of ordinary skill in the art recognizes that in all chemical compounds with a H atom, the H atom actually represents a mixture of H and D, with about 0.015% being D. Thus, compounds with a level of deuterium that has been enriched to be greater than its natural abundance of 0.015%, should be considered unnatural and, as a result, novel over their nonenriched counterparts. Thus, the present invention relates to a deuterium-enriched compound or compounds whose enrichment is greater than naturally occurring deuterated molecules.
[0045] All percentages given for the amount of deuterium present are mole percentages. Further, when a variable is not accompanied by a definition, the previous definition of the variable controls.
[0046] Unless indicated otherwise, when a D is specifically recited at a position or is shown in a formula, this D represents a mixture of hydrogen and deuterium where the amount of deuterium is about 100% (i.e., the abundance of deuterium is from 80% to 100%). In certain aspects, the abundance of deuterium is from 97% to 100%.
[0047] Compound refers to a quantity of molecules that is sufficient to be weighed, tested for its structural identity, and to have a demonstrable use (e.g., a quantity that can be shown to be active in an assay, an in vitro test, or in vivo test, or a quantity that can be administered to a patient and provide a therapeutic benefit).
[0048] As used herein, “lenalidomide” refers to a compound having the formula (II):
Figure imgf000012_0001
or pharmaceutically acceptable salts thereof. Such salts may include, but are not limited to, lenalidomide benzenesulfonate and lenalidomide p-toluenesulfonate. Lenalidomide may be referred to by other names, for example, its IUPAC name: 3-(4-amino-l-oxoisoindolin-2- yl)piperidine-2, 6-dione, its CAS number: 191732-72-6, Revimid, or its common brand name: Revlimid®. [0049] As used herein, “rituximab” refers to a chimeric monoclonal antibody targeted against CD20. Rituximab may be referred to by other names, for example, its CAS number: 174722-31-
7, or its common brand names: Rituxan®, Ruxience®, Truxima®, and MabThera®.
[0050] As used herein, “tazemetostat” refers to a compound of formula (III):
Figure imgf000013_0001
or pharmaceutically acceptable salts thereof. Such salts may include, but are not limited to, tazemetostat hydrobromide. Tazemetostat may be referred to by other names, for example, its IUPAC name: A-[(4,6-dimethyl-2-oxo-177-pyridin-3-yl)methyl]-3-[ethyl(oxan-4-yl)amino]-2- methyl-5-[4-(morpholin-4-ylmethyl)phenyl]benzamide, its CAS number: 1403254-99-8, EPZ- 6438, or its common brand name: Tazverik®.
[0051] As used herein, “venetoclax” refers to a compound of formula (IV):
Figure imgf000013_0002
or pharmaceutically acceptable salts thereof. Such salts may include, but are not limited to, venetoclax docusate. Venetoclax may be referred to by other names, for example, its IUPAC name: 4-[4-[[2-(4-chlorophenyl)-4,4-dimethylcyclohexen-l-yl]methyl]piperazin-l-yl]-7V-[3- nitro-4-(oxan-4-ylmethylamino)phenyl]sulfonyl-2-(177-pyrrolo[2,3-b]pyridin-5- yloxy)benzamide, its CAS number: 1257044-40-8, or its common brand names: Venclexta® and Venclyxto®. I. Compound of Formula (I)
[0052] The compound of formula (I), as depicted below, is a deuterium-enriched compound, and is also known as (N)-3-(5-amino-2-methyl-4-oxoquinazolin-3(4//)-yl)-(3-2H)-piperidine-2,6- dione:
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof.
[0053] The compound of formula (I) may also be referred to as “SP-3164” or “deuterated S- avadomide” throughout the present disclosure. A method of chemically synthesizing the compound of formula (I) (including Example 1 provided herein) is described in U.S. Patent No. 9,540,340, which is incorporated by reference in its entirety herein.
[0054] In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 85% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 90% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 95%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 90%.
[0055] The compound of formula (I) may also be characterized according to its stereochemical purity. The stereochemical purity of a compound containing one stereocenter can be characterized as enantiomeric excess (ee). Enantiomeric excess can be calculated using the formula: ee (%) = (major enantiomer - minor enantiomer)/(major enantiomer + minor enantiomer)* 100 where major and minor enantiomers are the most and least abundant enantiomer in a mixture of (R) and (S) enantiomers. [0056] In various embodiments, the compound of formula (I) has an enantiomeric excess of at least 5%.
[0057] In certain embodiments, the compound for formula (I) has an enantiomeric excess of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%. Additional examples of the stereoisomeric purity include an enantiomeric excess of at least about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about 70%, about 71%, about 72%, about 73%, about 74%, about 75%, about 76%, about 77%, about 78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99%.
[0058] In certain embodiments, the compound of formula (I) has a stereomeric purity (enantiomeric excess) of at least 5%.
[0059] In certain embodiments, the compound of formula (I) has a stereomeric purity (enantiomeric excess) of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%. Additional examples of the stereoisomeric purity (enantiomeric excess) of the compound of formula (I) include at least about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about
70%, about 71%, about 72%, about 73%, about 74%, about 75%, about 76%, about 77%, about
78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about
86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about
94%, about 95%, about 96%, about 97%, about 98%, or about 99%. In certain embodiments, the compound of formula (I) has an optical purity of at least 85% enantiomeric excess, 90% enantiomeric excess, 95% enantiomeric excess, or 98% enantiomeric excess.
[0060] In one aspect, provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent (e.g., rituximab, tazemetostat, or venetoclax) for the treatment of a lymphoma (e.g., follicular lymphoma (FL) or diffuse large B-cell lymphoma (DLBCL)) in a subject in need thereof.
[0061] In various embodiments, provided herein is a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent (e.g., rituximab, tazemetostat, or venetoclax) for the treatment of a lymphoma (e.g., FL or DLBCL) in a subject in need thereof.
[0062] In various embodiments, provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab for the treatment of DLBCL in a subject in need thereof.
[0063] In various embodiments, provided herein is a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab for the treatment of DLBCL in a subject in need thereof.
[0064] In various embodiments, provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat for the treatment of FL in a subject in need thereof.
[0065] In various embodiments, provided herein is a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat for the treatment of FL in a subject in need thereof.
[0066] In various embodiments, provided herein is a method of administering a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax for the treatment of FL in a subject in need thereof. [0067] In various embodiments, provided herein is a method of administering an effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax for the treatment of FL in a subject in need thereof.
III. Pharmaceutical Compositions
[0068] Provided herein are pharmaceutical compositions generally comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
[0069] In one aspect, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) a second therapeutic agent (e.g., rituximab, tazemetostat, or venetoclax) for the treatment of a lymphoma (e.g., follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL)) in a subject in need thereof.
[0070] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) an effective amount of a second therapeutic agent (e.g., rituximab, tazemetostat, or venetoclax) for the treatment of a lymphoma (e.g., FL or DLBCL) in a subject in need thereof.
[0071] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) rituximab for the treatment of DLBCL in a subject in need thereof.
[0072] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) an effective amount of rituximab for the treatment of DLBCL in a subject in need thereof.
[0073] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) tazemetostat for the treatment of FL in a subject in need thereof. [0074] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) an effective amount of tazemetostat for the treatment of FL in a subject in need thereof.
[0075] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) venetoclax for the treatment of FL in a subject in need thereof. [0076] In various embodiments, provided herein is a method of administering a combination of (i) a pharmaceutical composition comprising an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and (ii) an effective amount of venetoclax for the treatment of FL in a subject in need thereof.
[0077] The pharmaceutical compositions described herein can be administered by a variety of routes including, but not limited to, oral (enteral) administration, parenteral (by injection) administration, rectal administration, transdermal administration, intradermal administration, intrathecal administration, subcutaneous (SC) administration, intravenous (IV) administration, intramuscular (IM) administration, and intranasal administration. In certain embodiments, the pharmaceutical compositions described herein are administered orally.
[0078] The pharmaceutical compositions described herein may also be administered chronically (“chronic administration”). Chronic administration refers to administration of a compound or pharmaceutical composition thereof over an extended period of time, e.g., for example, over 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may be continued indefinitely, for example, for the rest of the subject’s life. In certain embodiments, the chronic administration is intended to provide a constant level of the compound in the blood, e.g., within the therapeutic window over the extended period of time.
[0079] The pharmaceutical compositions described herein may be presented in unit dosage forms to facilitate accurate dosing. The term “unit dosage forms” refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
[0080] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with ordinary experimentation. General considerations in the formulation and/or manufacture of pharmaceutical compositions can be found, for example, in Remington : The Science and Practice of Pharmacy 21 st ed. , Lippincott Williams & Wilkins, 2005.
III. Methods of Use and Treatment
[0081] In one aspect, provided herein are methods of treating a lymphoma in a subject in need thereof.
[0082] In various embodiments, provided herein is a method of treating a lymphoma in a subject in need thereof, comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful for the treatment of lymphomas.
[0083] In various embodiments, provided herein is a method of treating a lymphoma in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent useful for the treatment of lymphomas.
[0084] In certain embodiments, the second therapeutic agent useful for the treatment of lymphoma is selected from the group consisting of rituximab, tazemetostat, and venetoclax. [0085] In various embodiments, provided herein is a method of treating a lymphoma in a subject in need thereof, comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax.
[0086] In various embodiments, provided herein is a method of treating a lymphoma in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax. [0087] In certain embodiments, the lymphoma is follicular lymphoma (FL) or diffuse large B- cell lymphoma (DLBCL). In certain embodiments, the lymphoma is FL. In certain embodiments, the lymphoma is DLBCL.
[0088] In certain embodiments, the second therapeutic agent is rituximab. In certain embodiments, the second therapeutic agent is tazemetostat. In certain embodiments, the second therapeutic agent is tazemetostat.
[0089] In certain embodiments, the method reduces tumor volume or the rate of tumor growth in the subject. In certain embodiments, the method reduces tumor volume in the subject. In certain embodiments, the method reduces the rate of tumor growth in the subject.
[0090] In certain embodiments, the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of the second therapeutic agent. In certain embodiments, the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a 3) subject receiving only the effective amount of the second therapeutic agent. In certain embodiments, the method reduces the tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of the second therapeutic agent.
[0091] In certain embodiments, the second therapeutic agent is administered orally. In certain embodiments, the second therapeutic agent is administered intravenously. In certain embodiments, the second therapeutic agent is administered by intravenous infusion.
[0092] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat. [0093] In certain embodiments, the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat. In certain embodiments, the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat. In certain embodiments, the method reduces tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of tazemetostat.
[0094] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, the method comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax.
[0095] In certain embodiments, the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax. In certain embodiments, the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax. In certain embodiments, the method reduces tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of venetoclax.
[0096] In various embodiments, provided herein is a method of treating DLBCL in a subject in need thereof, the method comprising administering to the subject an effective amount of a combination of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab.
[0097] In certain embodiments, the method reduces the rate of tumor growth or tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab. In certain embodiments, the method reduces the rate of tumor growth in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab. In certain embodiments, the method reduces tumor volume in the subject below that of 1) a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, 2) a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or 3) a subject receiving only the effective amount of rituximab.
[0098] In certain embodiments, the tazemetostat is administered orally. In certain embodiments, the venetoclax is administered orally. In certain embodiments, the rituximab is administered intravenously. In certain embodiments, the rituximab is administered by intravenous infusion. [0099] In certain embodiments, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered orally.
[00100] In certain embodiments, the compound of formula (I) is administered in its free base form. In certain embodiments, the compound of formula (I) is administered as a pharmaceutically acceptable salt. [00101] In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 85% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 90% to about 100%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 95%. In certain embodiments, the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 90%.
[00102] In various embodiments, provided herein is a method of treating a lymphoma (e.g., FL or DLBCL) in a subject in need thereof, comprising administering to the subject a pharmaceutical composition described herein and a second therapeutic agent (e.g., rituximab, tazemetostat, and venetoclax) useful in the treatment of lymphoma.
[00103] In various embodiments, provided herein is a method of treating a lymphoma (e.g., FL or DLBCL) in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and a second therapeutic agent (e.g., rituximab, tazemetostat, and venetoclax) useful in the treatment of lymphoma.
[00104] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject a pharmaceutical composition described herein and tazemetostat.
[00105] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and tazemetostat.
[00106] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject a pharmaceutical composition described herein and venetoclax.
[00107] In various embodiments, provided herein is a method of treating FL in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and venetoclax. [00108] In various embodiments, provided herein is a method of treating DLBCL in a subject in need thereof, comprising administering to the subject a pharmaceutical composition described herein and rituximab.
[00109] In various embodiments, provided herein is a method of treating DLBCL in a subject in need thereof, comprising administering to the subject an effective amount of a combination of a pharmaceutical composition described herein and rituximab.
[00110] Certain combinations described herein (e.g., a compound of formula (I) combined with rituximab, tazemetostat, or venetoclax) may work synergistically in the treatment of the diseases, disorders, and conditions described herein (e.g., a lymphoma) and the symptoms associated with such diseases or disorders.
EXAMPLES
[00111] In order that the disclosure described herein may be more fully understood, the following examples are set forth. The examples described in this application are offered to illustrate the compositions and methods provided herein and are not to be construed in any way as limiting their scope.
Definitions
Abbreviations Full-text & descriptions
HPLC High-performance liquid chromatography
NMR Nuclear Magnetic Resonance Spectroscopy
MS Mass spectrometry
MeOH Methanol
DMSO Dimethyl sulfoxide
ROA Route of administration
PO Oral (ly)
IV Intravenous (ly)
BID Twice a day
BIW Twice a week
QD Once a day
BW Body weight BWL Body weight loss
IACUC Institutional animal care and use committee
RTV Relative tumor volume
TV Tumor volume
TGI Tumor Growth Inhibition; TGI% = (1-Ti/Ci) x 100;
Ti and Ci are the mean tumor volumes of the treatment and control groups, respectively, on a given day.
T/C The Ti/Ci value (%): T/C% = Ti/Ci x 100;
Ti and Ci are the mean tumor volumes of the treatment and control groups, respectively, on a given day.
Example 1. Synthesis of rac-3-(5-Amino-2-methyl-4-oxoquinazolin-3(47f)-yl)-(3-2H)- piperidine-2, 6-dione.
Step 1. Synthesis of 2-Methyl-5-nitro-4H-3,l-benzoxazin-4-one.
Figure imgf000025_0001
[00112] 2-Amino-6-nitrobenzoic acid (25.0 g, 137 mmol) was mixed with acetic anhydride (50 mL, 529 mmol) and the resulting mixture was heated to 120 °C by placing the reaction vessel containing this mixture in an oil bath for 2 hours, while monitoring reaction progress by HPLC. Next, the reaction mixture was cooled, and partially concentrated by evaporation under a stream of dry nitrogen. The reaction mixture was then diluted with 100 mL diethyl ether and cooled in a refrigerator overnight. The resulting crystals were filtered and rinsed with diethyl ether (50 mL) to provide the title compound (25.3 g, 123 mmol, 89%) as a tan, crystalline solid. !H NMR (400 MHz, CDCh) 5 7.91 (t, J = 8.0 Hz, 1H), 7.57 (dd, J = 7.8, 1.01 Hz, 1H), 7.53 (dd, J = 8.3, 1.0 Hz, 1H), 2.53 (s, 3H). MS (ESI+) calc, for [C9H6N2O4+H]+ 207.2, found 207.2.
Step 2. Synthesis of 2-Acetamido-6-nitrobenzoic acid
Figure imgf000025_0002
[00113] The starting material (24.7 g, 120 mmol) was dispersed in water (216 mL) and the resulting mixture was heated to reflux for 30 min. Upon cooling the reaction mixture, crystallization began. Next, the reaction mixture was placed in a refrigerator for 18 hours to complete crystallization. The crystalline material was isolated by filtration and dried under vacuum to give the title compound (24.3 g, 108 mmol, 90.5%) as a pale yellow, crystalline solid. 'H NMR (300 MHz, MeOH-d4) 5 8.49 (d, J = 8.1 Hz, 1H), 8.15 (d, J = 7.8 Hz, 1H), 8.01 (t, J = 8.2 Hz, 1 H), 2.53 (s, 3H). MS (ESI-) calc, for [CU^Cb-H]’ 223.0, found 223.1.
Step 3. Synthesis of rac-2-Acetamido-N-(2, 6-dioxopiperidin-3-yl)-6-nitrobenzamide
Figure imgf000026_0001
[00114] The starting acid (3.10 g, 13.8 mmol) was mixed with hydroxybenzotriazole (HOBt, 2.12 g of the hydrate, 13.8 mmol) and l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC, 2.54 g, 13.3 mmol), under a nitrogen atmosphere. N,N-dimethylformamide (DMF, 21.4 mL) was added and the mixture was stirred for 30 minutes at room temperature, rac- 3-Aminopiperidine-2, 6-dione hydrochloride (5.01 g, 30.4 mmol) was added, followed by N,N- diisopropylethylamine (DIEA, 9.63 mL, 55.3 mmol). The reaction mixture was stirred at 20 °C, while monitoring by HPLC. After 24 hours, the reaction mixture showed approximately 40% conversion to the desired product containing some remaining starting acid, but no amine. Then, the reaction mixture was slowly poured into 200 mL water with vigorous stirring. After 20 minutes, a white precipitate began to form. The mixture was placed in the refrigerator for 18 hours. Then, the precipitate was isolated by filtration. The filter cake was washed with 50 mL ether, and air dried to provide the title compound (1.60 g, 4.79 mmol, 35%) as a white powder. XH NMR (300 MHz, DMSO-d6) 5 11.16 (s, 1H), 9.40 (s, 1H), 9.34 (d, J = 8.0 Hz, 1H), 8.53 (d, J = 7.5 Hz, 1H), 7.89 (dd, J = 8.2, 0.98 Hz, 1H), 7.66 (t, J = 8.3 Hz, 1H), 4.79 (m, 1H), 2.85(m, 1H), 2.59 (m, 1H), 2.21 (m, 1H), 2.20 (s, 3H), 2.03 (m, 1H). MS (ESI-) calc, for [C14H14N4O6- H]’ 333.3, found 333.2. Step 4. Synthesis of rac-3-(2-Methyl-5-nitro-4-oxoquinazolin-3(4H)-yl)-(3-2H)-piperidine-2,6- dione
Figure imgf000027_0001
[00115] rac-2-Acetamido-N-(2,6-dioxopiperidin-3-yl)-6-nitrobenzamide (4.50 g, 13.5 mmol) was dispersed in anhydrous acetonitrile (70.3 mL) under a nitrogen atmosphere. Triethylamine (EtsN, 88.2 mL, 633 mmol) was added via syringe, followed by the dropwise addition of chlorotrimethylsilane (TMSCI, 25.6 mL, 202 mmol). The reaction mixture was warmed to 75 °C and the reaction was monitored by HPLC. After 42 hours, 85% of the desired product was present. The reaction mixture was cooled to 20 °C, stirred rapidly, and quenched with deuterium oxide (100 mL, 5.55 moles). The reaction mixture was stirred for an additional 20 minutes and a white precipitate formed. Next, the reaction mixture was cooled in a refrigerator for 4 hours and filtered to provide the title compound (2.25 g, 7.1 mmol, 53%) as an off-white, crystalline solid. 1H NMR (300 MHz, DMSO-d6) 5 11.07 (s, 1H), 7.98 (t, J = 7.6 Hz, 1H), 7.83 (2 dd, J = 14.4, 8.0, 1.2 Hz, 2H), 2,82 (m, 1H), 2.67 (s, 3H), 2.56 (m, 2H), 2.18 (m, 1H). MS (ESI-) calc, for [CI4HII[2H]N4O5-H]- 316.1, found 316.0.
Step 5. Synthesis of rac-3-(5-Amino-2-methyl-4-oxoquinazolin-3(4H)-yl)-(3-2H)-piperidine-2,6- dione
Figure imgf000027_0002
[00116] 3-(2-Methyl-5-nitro-4-oxoquinazolin-3(477)-yl)-(3-2H)-piperidine-2, 6-dione (2.25 g, 7.09 mmol) was dispersed in DMF (60 mL) in a Parr bottle and palladium hydroxide (500 mg, 20% active catalyst, 50 wt% water) was added. The bottle was then placed under 50 psi hydrogen pressure and shaken for 2 hours at 21 °C. The reaction mixture was analyzed by HPLC, which showed a major peak for the desired product and no starting material. The reaction mixture was treated with activated carbon and filtered through a plug of silica gel overlain with 1 cm of sodium sulfate. The plug was washed with 50 mL acetonitrile and the filtrate was evaporated under high vacuum to give a black gum. This material was dissolved in 50 mL of acetonitrile and filtered through a Magnesol® plug, then washed with 100 mL of acetonitrile. [00117] The filtrate was placed in a freezer for 48 hours, and then filtered to afford smoky grey crystals, which were washed with diethyl ether (50 mL) and air dried for 3 hours to give the title compound (1.63 g, 5.67 mmol, 80%). ’H NMR (400 MHz, DMSO-d6) 5 10.99 (s, 1H), 7.37 (t, J = 8.1 Hz, 1H), 7.03 (br s, 2H), 6.58 (dd, J = 11.2, 8.1 Hz, 2H), 2.83 (m, 0.92H), 2.60 (m, 2H), 2.53 (s, 3H), 2.14 (m, 0.92H). MS (ESI+) calc, for [Ci4Hi3[2H]N4O3+H]+ 288.1, found 288.2. [00118] ’H NMR (400 MHz, pyndine-d5) 5 13.14 (br s, 1H), 7.69 (br s, 2 H), 7.46 (t, J = 8.1 Hz, 1H), 7.06 (dd, J = 7.8, 0.9 Hz, 1H), 6.78 (dd, J = 8.2, 0.9 Hz, 1H), 5.39 (dd, J = 11.5, 5.7 Hz, 0.06H, residual C(3)H, ca. 94% D incorporation], 3.12 (m, 1H), 2.94 [m, 1.81H, C(5)H, ca. 19% D incorporation], 2.17 (s, 3H), 2.19 (ddd, J = 3.2, 4.6, 12.6, 1H).
[00119] 'H NMR in pyridine-ds indicates approximately 94% deuterium incorporation at C(3), and approximately 19% deuterium incorporation at C(5). Mass spectroscopic analysis indicates 4% protonated, 80% mono-deuterated, and 16% bis-deuterated.
Example 2. Separation of Pure Enantiomers of rac-3-(5-Amino-2-methyl-4-oxoquinazolin- 3(4/f)-yl)-(3-2H)-piperidine-2, 6-dione
[00120] Separation of 0.948 g of rac-3-(5-amino-2-methyl-4-oxoquinazolin-3(477)-yl)-(3- 2H)piperidine-2, 6-dione (3.3 mmol) gave the two pure enantiomers in 726.7 mg (2.53 mmol, 77%) overall yield. It was achieved using the following method.
[00121] rac-3-(5-Amino-2-methyl-4-oxoquinazolin-3(4/ )-yl)-(3-2H)piperidine-2, 6-dione was dissolved in 30 mL of a mixture of acetonitrile, isopropanol, and methanol (5:3:2 v/v/v). The enantiomers were separated by chiral supercritical fluid chromatography on a ChiralPak AD-H column (21 x 250 mm) using a mobile phase of 37% isopropanol in carbon dioxide (flow rate: 70 mL/min; 1 mL injected per run). Compounds were detected by UV at 254 nm. Fractions containing the compounds were pooled and evaporated. Purity and enantiomeric excess (% ee = % enantiomer 1 - % enantiomer 2) were determined by analytical supercritical fluid chromatography on a ChiralPak AD-H column (4.6 x 100 mm) using the same eluent (sample concentration ~ 1 mg/mL in methanol; flow rate 3 mL/min). Deuterium content was measured by LC/MS-MS and optical rotation was measured in N,N-dimethylformamide (DMF) at room temperature.
[00122] (X)-3-(5-Amino-2-methyl-4-oxoquinazolin-3(4H)-yl)-(3-2H)piperidine-2, 6-dione:
340 mg (1.18 mmol); 99.8% purity; 99.6% ee; LC-MS: 288.3 (M+l) (90.3% deuterium);
Figure imgf000029_0001
NMR (200 MHz, DMSO-d6) 5 10.95 (s, 1H), 7.33 (t, J = 8 Hz, 1H), 6.99 (br s, 2H), 6.56 (d, J = 8 Hz, 1H), 6.52 (d, J = 6 Hz, 1H), 5.1 (m, 0.12H, residual C(3)H, approximately 88% deuterium incorporation), 2.75 (m, 6H), 2.11 (m, 1H); optical rotation [OC]D = -47.2°(c 1.0, 19.3 °C, DMF). [00123] (R)-3-(5-Amino-2-methyl-4-oxoquinazolin-3(4H)-yl)-(3-2H)piperidine-2, 6-dione: 386.7 mg (1.35 mmol); 99.2% purity; 98.4% ee; LC-MS: 288.3 (M+l) (92.6% deuterium); ’H NMR (300 MHz, DMSO-d6) 5 11.04 (s, 1H), 7.40 (t, J = 8.0 Hz, 1H), 7.0 (br s, 2H), 6.61 (d, J = 8.0, 2H), 5.2 (m, 0.14H, residual C(3)H, approximately 86% deuterium incorporation), 2.82 (m, 1H), 2.61 (m, 5H), 2.15 (m, 1H); optical rotation [a]D = +43.35°(c 1.0, 19.3 °C, DMF).
Example 3. In Vivo Efficacy Study of SP-3164 and Rituximab Combination in a Subcutaneous WSU-DLCL-2 Human Diffuse Large B Cell Lymphoma (DLBCL) Xenograft Model in Female NOD/SCID Mice
[00124] Female Mus musculus mice with an NOD/SCID genetic profile (6-8 weeks estimated average age at inoculation; Beijing Vital River Laboratory Animal Technology Co., Ltd) were used in the study.
[00125] Animals were housed in polysulfone individually ventilated cages (IVC) cages (335mm x 143mm x 130mm, up to 5 per cage), supplied with autoclaved crushed corncob bedding that was changed weekly. The temperature and humidity were maintained at 20 - 26 °C and 40 - 70% humidity. A light cycle of 12 hours light and 12 hours dark was used. Mice were fed standard rodent chow (irradiated) and autoclaved, filtered (reverse osmosis) softened water ad libitum. Each mouse was identified by a labeled ear tag. Cages were identified by label, which included project number, group number, number of animals, gender, strain, supplier, date of receipt, treatment, animal use protocol (AUP) number, and date of treatment initiation. Daily cage side and weekly clinical observations were made. The protocol was reviewed and approved by the IACUC. During the study, the care and use of animals was conducted in accordance with the regulations of the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC). [00126] The study was designed to compare treatment effects of lenalidomide, SP-3164, and rituximab monotherapies and to compare combination therapies of lenalidomide and rituximab and SP-3164 and rituximab in a WSU-DLCL2 DLBCL xenograft model. Lenalidomide and SP- 3164 were administered orally (PO) twice daily (BID with an interval of 5h between the two doses each day). Rituximab was given intravenously (IV) bi-weekly (BIW).
[00127] The study design and dose levels are provided in Table 1.
Table 1. Study Design for Dosing Frequency and Duration
Figure imgf000030_0001
[00128] Formulations were prepared in a biosafety cabinet. The dosing volume was adjusted for body weight (dosing volume = 10 mL/kg). The formulations are provided in Table 2.
Table 2. Study Drug Formulations
Figure imgf000030_0002
[00129] WSU-DLCL2 cells were maintained in vitro with RPMI-1640 medium supplemented with 10% fetal bovine serum at 37 °C in an atmosphere of 5% CO2 in air. Cells in exponential growth phase were harvested and quantitated by cell counter before tumor inoculation.
[00130] Each mouse was inoculated subcutaneously in the right upper flank region with WSU- DLCL2 cells (lx 107) in 0.1 ml of PBS mixed with Matrigel (1 : 1) for tumor development.
[00131] As shown in Table 1, animals were randomly allocated to 6 study groups, (N = 10 per group) when the mean tumor size reached approximately 150-200 mm3. Randomization was performed based on the “matched distribution” method (Study Director™ software, version 3.1.399.19). The date of randomization was denoted as Day 0.
[00132] Treatment was initiated on the day of randomization (Day 0) per study design. [00133] After tumor cells inoculation, the animals were checked daily for morbidity and mortality. During routine monitoring, the animals were checked for any effects of tumor growth and treatments on behavior such as mobility, food and water consumption, body weight gain/loss (body weights were measured twice per week after randomization), eye/hair matting and any other abnormalities. Mortality and observed clinical signs were recorded for individual animals. Tumor volumes were measured twice per week in two dimensions using a caliper, and the volume was expressed in mm3 using the formula V = (L x W x W)/2, where V is tumor volume, L is tumor length (the longest tumor dimension) and W is tumor width (the longest tumor dimension perpendicular to L). Dosing as well as tumor and body weight measurements were conducted in a laminar flow cabinet. The body weights and tumor volumes were recorded by using StudyDirector™ (version 3.1.399.19).
[00134] After one measurement of BWL > 15%, a dosing holiday was given to the individual mouse, which was then housed in a separate cage alone and monitored daily, while the other animals in the same group received dosing as scheduled. Treatment of the mouse with BWL > 15% resumed when body weight loss had recovered to BWL < 10%. Supplemental feeding (gel) was provided to all the mice in the same group, if an individual animal with BWL > 15% was observed in the group.
[00135] Additionally, animals were scheduled to be euthanized if they lost more than 20% of their body weight relative to their weight on the first day of treatment. Similarly, animals were to be euthanized if their tumor volume were to exceed 3000 mm3. To deter cannibalization, any animal exhibiting an ulcerated or necrotic tumor was immediately separated, singly housed, and monitored daily before euthanasia or until tumor regression was complete. If the tumor ulcerated, and the ulceration diameter was greater than 5 mm, or pus or necrosis was observed; or if the tumor burden, including metastasis, compromised an animal’s normal physiologic performances, (e.g., orientation, access to food or water), the animal was euthanized rapidly. Finally, a mouse was euthanized rapidly if its general welfare was deemed unacceptable (e.g., emaciated (skeletal structure extremely prominent, little or no flesh cover, vertebrae distinctly segmented); persistent hypothermia; blood-stained or mucopurulent discharge from any orifice; labored breathing, particularly accompanied by nasal discharge or cyanosis; hind limb paralysis or weakness (cannot reach food and water); severe anemia (mainly indicated by pale feet or ear); severe infection; or incontinence or diarrhea over 48 hours or severe dehydration).
[00136] Mouse treatment was performed for 28 days. Following cessation of treatment, animals and tumor growth were monitored until a humane termination endpoint was reached.
[00137] TGI analysis where the calculation was based on absolute tumor volume and relative tumor volume (RTV) was applied to the final data interpretation. T/C analysis on the absolute T/C ratio and relative T/C ratio (RTV) was applied to the final data interpretation. Survival (Kaplan-Meier model with log-rank method) analysis was applied to the final data interpretation. To compare tumor volumes differences amongst groups unpaired, two-tailed Student’s ttests were performed and p values <0.05 regarded as statistically significant.
[00138] The activity of SP-3164 in combination with rituximab was compared to that of lenalidomide in combination with rituximab in the mouse WSU-DLCL2 DLBCL xenograft model. Mice were treated for 28 days and tumor volume was measured twice weekly. The combination of SP-3164 and rituximab resulted in sustained regressions and complete responses (tumor = 0 mm3) in 50% of treated mice (FIG. 1). Greater TGI was observed for SP-3164 as compared to lenalidomide and greater antitumor activity for the combination of SP-3164 and rituximab as compared to either SP-3164 or rituximab alone or to the combination of lenalidomide and rituximab. Example 4. In Vivo Efficacy Study of SP-3164 and Tazemetostat and SP-3164 and Venetoclax Combinations in a Subcutaneous DOHH-2 Human B-Cell Follicular Lymphoma Xenograft Model in Female CB17/SCID Mice
[00139] The study was performed in female Mus musculus mice with a CB17/SCID genetic profile, an estimated average age and body weight at inoculation of 6-8 weeks, and > 16 g, respectively (mice from the Beijing Vital River Laboratory Animal Technology Co., Ltd). [00140] Animals (up to 5 per cage) were housed in polysulfone IVC cages (335mm x 143 mm x 130mm) at 20 - 26 °C with 40 - 70% humidity, supplied with autoclaved crushed corncob bedding that was changed weekly, and kept on a light cycle of 12 hours light and 12 hours dark. Mice were fed standard rodent chow (irradiated) and autoclaved filtered (reverse osmosis) softened water ad libitum. Each mouse was identified by a labeled ear tag. Cages were identified by label, which included the project number, group number, number of animals, gender, strain, supplier, date of receipt, treatment, AUP number, and date of treatment initiation. Cage sides were monitored daily in conjunction with weekly clinical observations.
[00141] The study was designed to compare the treatment effect of SP-3164, tazemetostat and venetoclax alone or in combinations (SP-3164 and tazemetostat or SP-3164 and venetoclax only) in a DOHH-2 xenograft model of FL. Dosing groups, dose levels, and dosing schedule are provided in Table 3.
Table 3. Study Design for Dosing Frequency and Duration (aa compounds administered orally).
Figure imgf000033_0001
[00142] Formulations (Table 4) were prepared in a biosafety cabinet. The dosing volume was adjusted for body weight (dosing volume = 10 mL/kg).
Table 4. Study Drug Formulations
Figure imgf000034_0001
[00143] The protocol of the study was reviewed and approved by the IACUC. During the study, the care and use of animals was conducted in accordance with the regulations of the AAALAC.
[00144] DOHH-2 cells were maintained in vitro with RPMI1640 medium supplemented with 10% fetal bovine serum at 37 °C in an atmosphere of 5% CO2 in the air. Cells in exponential growth phase were harvested and quantitated by cell counter before tumor inoculation.
[00145] Each mouse was inoculated subcutaneously in the right upper flank region with DOHH-2 cells (5 x 106) in 0.1 ml of PBS mixed with Matrigel (1 :1) for tumor development.
[00146] Animals were randomly allocated to 6 study groups, (N = 10 per group) when the mean tumor size reached approximately 80-150 mm3 (see Table 3). Randomization was performed based on the “matched distribution” method (StudyDirector™ software, version 3.1.399.19). The date of randomization was denoted as Day 0.
[00147] Treatment was initiated on Day 0.
[00148] After tumor cells inoculation, the animals were checked daily for morbidity and mortality. During routine monitoring, the animals were checked for any effects of tumor growth and treatments on behavior such as mobility, food and water consumption, body weight gain/loss (body weights will be measured twice per week after randomization), eye/hair matting and any other abnormalities. Mortality and observed clinical signs were recorded for individual animals in detail. Tumor volumes were measured twice weekly after randomization in two dimensions using a caliper, and the volume expressed in mm3 was calculated using the formula V = (L x W x W)/2, where V is tumor volume, L is tumor length (the longest tumor dimension) and W is tumor width (the longest tumor dimension perpendicular to L). Dosing as well as tumor and body weight measurements were performed in a laminar flow cabinet. The body weights and tumor volumes were recorded using the StudyDirector™ software (version 3.1.399.19). Criteria for dosing holidays, supplemental gel administration, and humane termination were the same as for the WSU-DLCL2 DLBCL xenograft experiment described in Example 3. Drug treatment was performed for 28 days. The data was statistically analyzed as described above for the WSU- DLCL2 DLBCL xenograft model (Example 3).
[00149] FIG. 2 shows that SP-3164 (7.5 mg/kg BID PO) demonstrated significant tumor growth inhibition (TGI) compared to vehicle and out-performed tazemetostat monotherapy (300 mg/kg BID PO). A synergistic effect on TGI was observed for the combination of SP-3164 and tazemetostat when compared to SP-3164 alone. This suggests that SP-3164 may sensitize FL cells to tazemetostat.
[00150] FIG. 3 shows that SP-3164 (7.5 mg/kg BID PO) showed significant TGI compared to vehicle. The combination of SP-3164 and venetoclax demonstrated a synergistic TGI effect when compared to venetoclax or SP-3164 alone.
INCORPORATION BY REFERENCE
[00151] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.
EQUIVALENTS
[00152] The disclosure may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the disclosure described herein. Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

WHAT IS CLAIMED:
1. A method of treating a lymphoma in a subject in need thereof, the method comprising administering to the subject an effective amount of a combination of a compound of formula (I)
Figure imgf000036_0001
or a pharmaceutically acceptable salt thereof, and a second therapeutic agent selected from the group consisting of rituximab, tazemetostat, and venetoclax.
2. The method of claim 1, wherein the lymphoma is follicular lymphoma or diffuse large B-cell lymphoma.
3. The method of claim 1 or 2, wherein the lymphoma is follicular lymphoma.
4. The method of any one of claims 1-3, wherein the second therapeutic agent is tazemetostat.
5. The method of any one of claims 1-3, wherein the second therapeutic agent is venetoclax.
6. The method of claim 1 or 2, wherein the lymphoma is diffuse large B-cell lymphoma.
7. The method of any one of claims 1 , 2, and 6, wherein the second therapeutic agent is rituximab.
8. The method of any one of claims 1-7, wherein the method reduces tumor growth or volume in the subject.
9. The method of any one of claims 1-8, wherein the method reduces tumor growth or volume in the subject below that of a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the second therapeutic agent, or that of a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or that of a subject receiving only the effective amount of the second therapeutic agent.
10. A method of treating follicular lymphoma in a subject in need thereof, the method comprising administering to the subject an effective amount of a combination of a compound of formula (I)
Figure imgf000037_0001
or a pharmaceutically acceptable salt thereof, and tazemetostat.
11. The method of claim 10, wherein the method reduces tumor growth or volume in the subject below that of a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and tazemetostat, or that of a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or that of a subject receiving only the effective amount of tazemetostat.
12. A method of treating follicular lymphoma in a subject in need thereof, the method comprising administering to the subject an effective amount of a combination of a compound of formula (I)
Figure imgf000038_0001
or a pharmaceutically acceptable salt thereof, and venetoclax.
13. The method of claim 12, wherein the method reduces tumor growth or volume in the subject below that of a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and venetoclax, or that of a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or that of a subject receiving only the effective amount of venetoclax.
14. A method of treating diffuse large B-cell lymphoma in a subject in need thereof, the method comprising administering to the subject an effective amount of a combination of a compound of formula (I)
Figure imgf000038_0002
15. The method of claim 14, wherein the method reduces tumor growth or volume in the subject below that of a subject not receiving the effective amount of a combination of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and rituximab, or that of a subject receiving only the effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or that of a subject receiving only the effective amount of rituximab.
16. The method of any one of claims 1-15, wherein the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered orally.
17. The method of any one of claims 1-16, wherein the compound of formula (I) is administered in its free base form.
18. The method of any one of claims 1-17, wherein the abundance of deuterium at the position labeled D in the compound of formula (I) is about 80% to about 100%.
PCT/US2023/080129 2022-11-16 2023-11-16 Methods of use of deuterium-enriched compounds WO2024108028A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263425898P 2022-11-16 2022-11-16
US63/425,898 2022-11-16

Publications (1)

Publication Number Publication Date
WO2024108028A1 true WO2024108028A1 (en) 2024-05-23

Family

ID=89190699

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/080129 WO2024108028A1 (en) 2022-11-16 2023-11-16 Methods of use of deuterium-enriched compounds

Country Status (1)

Country Link
WO (1) WO2024108028A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014110558A1 (en) * 2013-01-14 2014-07-17 Deuterx, Llc 3-(5-substituted-4-oxoquinazolin-3(4h)-yl)-3-deutero-piperidine-2,6-dione derivatives
WO2016007854A1 (en) * 2014-07-11 2016-01-14 Celgene Corporation Combination therapy for cancer
WO2017096024A1 (en) * 2015-12-02 2017-06-08 Celgene Corporation Cycling therapy using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014110558A1 (en) * 2013-01-14 2014-07-17 Deuterx, Llc 3-(5-substituted-4-oxoquinazolin-3(4h)-yl)-3-deutero-piperidine-2,6-dione derivatives
US9540340B2 (en) 2013-01-14 2017-01-10 Deuterx, Llc 3-(5-substituted-4-oxoquinazolin-3(4H)-yl)-3-deutero-piperidine-2,6-dione derivatives and compositions comprising and methods of using the same
WO2016007854A1 (en) * 2014-07-11 2016-01-14 Celgene Corporation Combination therapy for cancer
WO2017096024A1 (en) * 2015-12-02 2017-06-08 Celgene Corporation Cycling therapy using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
ANONYMOUS: "SP-3164, a cereblon-binding molecular glue", 26 October 2022 (2022-10-26), pages 1 - 26, XP093132834, Retrieved from the Internet <URL:https://investors.salariuspharma.com/static-files/66f971cb-ff14-4035-af74-8fbebcef5f32> [retrieved on 20240219] *
MARTIN: "Remington's Pharmaceutical Sciences", 1975, MACK PUBL. CO.
NASTOUPIL LORETTA J ET AL: "Avadomide (CC-122), a Novel Cereblon Modulating Agent, Plus Rituximab in Patients with Relapsed or Refractory Follicular Lymphoma", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 132, 29 November 2018 (2018-11-29), pages 1602, XP086595507, ISSN: 0006-4971, DOI: 10.1182/BLOOD-2018-99-114409 *
NASTOUPIL LORETTA J. ET AL: "Phase Ib study of avadomide (CC-122) in combination with rituximab in patients with relapsed/refractory diffuse large B-cell lymphoma and follicular lymphoma", EJHAEM, vol. 3, no. 2, 1 May 2022 (2022-05-01), pages 394 - 405, XP093133280, ISSN: 2688-6146, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9175947/pdf/JHA2-3-394.pdf> DOI: 10.1002/jha2.394 *
RIBRAG VINCENT ET AL: "Phase Ib study of combinations of avadomide (CC-122), CC-223, CC-292, and rituximab in patients with relapsed/refractory diffuse large B-cell lymphoma", EJHAEM, vol. 3, no. 1, 14 January 2022 (2022-01-14), pages 139 - 153, XP093133502, ISSN: 2688-6146, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1002/jha2.375> DOI: 10.1002/jha2.375 *
SANTIESTEBAN DANIELA Y ET AL: "SP-3164, a Novel Cereblon-Binding Protein Degrader, Shows Activity in Preclinical Lymphoma Models", BLOOD, vol. 140, no. Supplement 1, 11 December 2022 (2022-12-11), US, pages 8865 - 8866, XP093133522, ISSN: 0006-4971, DOI: 10.1182/blood-2022-167495 *
SANTIESTEBAN DANIELA Y ET AL: "SP-3164, a Novel Cereblon-Binding Protein Degrader, Shows Activity in Preclinical Lymphoma Models", BLOOD, vol. 140, no. Supplement 1, 15 November 2022 (2022-11-15), US, pages 8865 - 8866, XP093132843, ISSN: 0006-4971, Retrieved from the Internet <URL:https://ashpublications.org/blood/article/140/Supplement%201/8865/492443/SP-3164-a-Novel-Cereblon-Binding-Protein-Degrader> DOI: 10.1182/blood-2022-167495 *
SANTIESTEBAN DANIELA Y ET AL: "SP-3164, a novel Ikaros and Aiolos molecular glue degrader with preclinical activity in non-Hodgkin lymphomas", 1 April 2023 (2023-04-01), pages 1 - 1, XP093133131, Retrieved from the Internet <URL:https://investors.salariuspharma.com/static-files/70a464be-4c53-4abd-abb1-c364bb76947b> [retrieved on 20240220] *
SANTIESTEBAN DANIELA Y. ET AL: "Abstract 524: SP-3164, a novel ikaros and aiolos molecular glue degrader with preclinical activity in non-Hodgkin lymphomas", CANCER RESEARCH, vol. 83, no. 7_Supplement, 4 April 2023 (2023-04-04), US, pages 524 - 524, XP093133133, ISSN: 1538-7445, Retrieved from the Internet <URL:https://aacrjournals.org/cancerres/article/83/7_Supplement/524/721115/Abstract-524-SP-3164-a-novel-ikaros-and-aiolos> DOI: 10.1158/1538-7445.AM2023-524 *

Similar Documents

Publication Publication Date Title
AU2017286370B2 (en) Heterocyclic compounds as antibacte rials
US20200239434A1 (en) Antibacterial compounds
JP2022066289A (en) Cannabinoid receptor modulators
US10017495B2 (en) Therapeutically active compounds and their methods of use
JP5897566B2 (en) Cyclic N, N&#39;-diarylthiourea and N, N&#39;-diarylurea-androgen receptor antagonists, anticancer agents, methods for their preparation and uses
TW201843136A (en) Therapeutically active compositions and their methods of use
TWI780061B (en) Uses of indolinone compounds
JP7078555B2 (en) Small molecule regulator of pantothenate kinase
JP6528779B2 (en) Preventive and / or therapeutic agent for diffuse large B cell lymphoma
EA031041B1 (en) 2,3,4,5-tetrahydropyridin-6-amine and 3,4-dihydro-2h-pyrrol-5-amine compound inhibitors of beta-secretase
EP2994457B1 (en) Radiomitigating pharmaceutical formulations
US20220380338A1 (en) Cycloalkyl 3-oxopiperazine carboxamides and cycloheteroalkyl 3-oxopiperazine carboxamides as nav1.8 inhibitors
US20120046302A1 (en) Methods of treating cns disorders
JPS62120317A (en) Agent for amelioration and remedy of dementia
CA3037766C (en) Dextrorphan-derivatives with suppressed central nervous activity
CA2905573C (en) A crystalline form of an anxiolytic compound
EA038510B1 (en) Indazole compounds for use in tendon and/or ligament injuries
WO2024108028A1 (en) Methods of use of deuterium-enriched compounds
EP4190770A1 (en) Dual regulator for mglur5 and 5-ht2a receptors and use thereof
US11299497B2 (en) Substituted tetrahydropyran dihydrothienopyrimidines and their use as phosphodiesterase inhibitors
CN108495852B (en) 2-amido thiazole derivatives, preparation method and application thereof
WO2024123991A1 (en) Treatment of autoimmune and inflammatory disorders with inhibitors of bet family bdii bromodomain
US20140275060A1 (en) Compounds for the treatment of neurologic disorders
OA19129A (en) Heterocyclic compounds as antibacterials.
TW202124359A (en) Methods and materials for increasing level of phosphorylated ampk protein