WO2017079003A1 - Composés deutérés pour le traitement de leucémies, compositions et procédés associés - Google Patents

Composés deutérés pour le traitement de leucémies, compositions et procédés associés Download PDF

Info

Publication number
WO2017079003A1
WO2017079003A1 PCT/US2016/058838 US2016058838W WO2017079003A1 WO 2017079003 A1 WO2017079003 A1 WO 2017079003A1 US 2016058838 W US2016058838 W US 2016058838W WO 2017079003 A1 WO2017079003 A1 WO 2017079003A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acid
idelalisib
compounds
Prior art date
Application number
PCT/US2016/058838
Other languages
English (en)
Inventor
Chaoran HUANG
Changfu CHENG
Original Assignee
NeuForm 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 NeuForm Pharmaceuticals, Inc. filed Critical NeuForm Pharmaceuticals, Inc.
Priority to CN201680060582.XA priority Critical patent/CN108137592B/zh
Publication of WO2017079003A1 publication Critical patent/WO2017079003A1/fr

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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine

Definitions

  • the invention generally relates to therapeutics and treatment methods for certain diseases and conditions. More particularly, the invention provides novel chemical compounds, including 5- fluoro-3-phenyl-2-[(lS)-l-(9H-purin-6-ylamino)propyl]quinazolin-4(3H)-one with deuterium- substitutions at strategic positions, useful for treating various blood cancers and related diseases and conditions, and pharmaceutical compositions and methods of preparation and use thereof.
  • Blood cancers are forms of cancer that begin in the cells of blood-forming tissue, for example, the bone marrow or in the cells of the immune system.
  • hematologic cancers include acute and chronic leukemias, lymphomas, multiple myeloma and myelodysplastic syndromes.
  • Leukemia is a group of cancers that usually begin in the bone marrow and result in high numbers of abnormal white blood cells.
  • Lymphocytic leukemia involves lymphocytes.
  • Myelogenous leukemia involves granulocytes. These white blood cells are important in fighting infections.
  • Lymphoma is a type of cancer that develops in the lymphatic system.
  • Leukemia can be of a type called acute leukemia, which is characterized by a rapid increase in the number of immature blood cells, making the bone marrow unable to produce healthy blood cells. Immediate treatment is needed to slow down the rapid progression and accumulation of the malignant cells, which can spread to other organs of the body via the bloodstream.
  • Leukemia can be of a type called chronic leukemia, for example, chronic lymphocytic leukemia (CLL), which is characterized by the excessive buildup of relatively mature, but still abnormal, white blood cells. Typically taking months or years to progress, the cells are produced at a much higher rate than normal, resulting in many abnormal white blood cells. Chronic forms are occasionally monitored for some time before treatment to ensure maximum effectiveness of therapy.
  • CLL chronic lymphocytic leukemia
  • lymphoma There are two general types of lymphoma depending on how cancer spreads. In Hodgkin lymphoma, the cancer spreads from one group of lymph nodes to another in a certain order. In non- Hodgkin lymphoma, the cancer spreads from one group of lymph nodes to another in a random order. Examples of non-Hodgkin lymphoma include follicular B-cell non-Hodgkin lymphoma (FL) and small lymphocytic lymphoma (SLL).
  • FL follicular B-cell non-Hodgkin lymphoma
  • SLL small lymphocytic lymphoma
  • Myeloma is a cancer that causes the plasma cells to form a tumor in the bone marrow. Myeloma is usually found in multiple places in the body, thus often called multiple myeloma.
  • the invention provides novel, orally available phosphoinositide 3-kinase (PI3K) delta inhibitors.
  • PI3K phosphoinositide 3-kinase
  • the chemical compounds and pharmaceutical compositions disclosed herein are biochemically potent and physiologically active with improved pharmacokinetic and toxicological properties over idelalisib.
  • the compounds disclosed herein are deuterium-substituted and improved versions of idelalisib, where hydrogen is substituted by deuterium at strategic locations of the molecule. The substitution locations are selected with the specific objective to impact
  • the resulting compounds have up to 7 deuterium substitutions and exhibit more desirable profiles in terms of safety, efficacy and tolerability in the treatment of blood cancers and related diseases and conditions, such as CLL, FL and SLL.
  • the invention generally relates to a compound having the structural formula of:
  • R 1; R 2 , R3, R4, R5, R6 and R7 is D
  • the invention generally relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound having the structural formula of:
  • each of R 1; R2, R3, R4, R5, R6 and R7 is D, or a pharmaceutically acceptable salt or ester thereof, effective to treat, prevent, or reduce one or more blood cancers, or a related disease or disorder thereof, in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier, or diluent.
  • the invention generally relates to a unit dosage form comprising the pharmaceutical composition disclosed herein.
  • the unit dosage is suitable for administration to a subject suffering from one or more blood cancers, including advanced hematologic malignancies, or a related disease or disorder thereof.
  • the invention generally relates to a method for treating, reducing, or preventing a disease or disorder.
  • the method includes: administering to a subject in need thereof a pharmaceutical composition comprising compound having the formula of:
  • each of R 1; R2, R 3 , R4, R5, R6 and R7 is D, or a pharmaceutically acceptable salt or ester thereof, effective to treat, prevent, or reduce one or more blood cancers, or related a related disease or disorder thereof.
  • the one or more blood cancers include diseases and conditions that may benefit from treatment using the compounds, pharmaceutical composition, unit dosage form and treatment method disclosed herein include any diseases and disorders that may be addressed through inhibition of the delta isoform of PI3K, such as ALL, FL and SLL.
  • the method of treatment includes administering to a subject in need thereof a pharmaceutical composition comprising compound having the formula of:
  • each of R 1; R2, R 3 , R4, R5, R6 and R7 is D, or a pharmaceutically acceptable salt or ester thereof, in combination with one or more other anti-cancer agents.
  • FIG. 1 shows certain exemplary 1 HNMR (CDCI 3 ) of a compound disclosed herein.
  • FIG. 2 shows certain exemplary 1 HNMR (CDCI 3 ) of a compound disclosed herein.
  • FIG. 3 shows certain exemplary LC-MS data of a compound disclosed herein.
  • FIG. 4 shows certain exemplary 1 HNMR (CDCI 3 ) of a compound disclosed herein.
  • FIG. 5 shows certain exemplary 1 HNMR (CDCI 3 ) of a compound disclosed herein.
  • FIG. 6 shows certain exemplary 1 HNMR (CDCI 3 ) of a compound disclosed herein.
  • FIG. 7 shows certain exemplary HPLC data of a compound disclosed herein.
  • FIG. 8 shows certain exemplary data on percentage of compound remaining vs. incubation time. After 4 hour, the concentration of D 7 -idelalisib is approximately equal to 220% of idelalisib.
  • FIG. 9 shows certain exemplary comparison of idelalisib vs. D 7- idelalisib in the formation of oxidative metabolites formed by purine ring oxidation on carbon-8.
  • administration encompasses the delivery to a subject of a compound as described herein, or a prodrug or other pharmaceutically acceptable derivative thereof, using any suitable formulation or route of administration, as discussed herein.
  • the terms "effective amount” or “therapeutically effective amount” refer to that amount of a compound or pharmaceutical composition described herein that is sufficient to effect the intended application including, but not limited to, disease treatment, as illustrated below.
  • the amount is that effective for detectable killing or inhibition of the growth or spread of cancer cells; the size or number of tumors; or other measure of the level, stage, progression or severity of the cancer.
  • the therapeutically effective amount can vary depending upon the intended application, or the subject and disease condition being treated, e.g., the desired biological endpoint, the pharmacokinetics of the compound, the disease being treated, the mode of administration, and the weight and age of the patient, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of cell migration.
  • the specific dose will vary depending on, for example, the particular compounds chosen, the species of subject and their age/existing health conditions or risk for health conditions, the dosing regimen to be followed, the severity of the disease, whether it is administered in combination with other agents, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • treatment or “treating” a disease or disorder refers to a method of reducing, delaying or ameliorating such a condition before or after it has occurred.
  • Treatment may be directed at one or more effects or symptoms of a disease and/or the underlying pathology. Treatment is aimed to obtain beneficial or desired results including, but not limited to, therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient can still be afflicted with the underlying disorder.
  • the a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient can still be afflicted with the underlying disorder.
  • compositions can be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • the treatment can be any reduction and can be, but is not limited to, the complete ablation of the disease or the symptoms of the disease. As compared with an equivalent untreated control, such reduction or degree of prevention is at least 5%, 10%, 20%, 40%, 50%, 60%, 80%, 90%, 95%, or 100% as measured by any standard technique.
  • the term "therapeutic effect” refers to a therapeutic benefit and/or a prophylactic benefit as described herein.
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any
  • esters refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Such esters can act as a prodrug as defined herein.
  • Pharmaceutically acceptable esters include, but are not limited to, alkyl, alkenyl, alkynyl, aryl, aralkyl, and cycloalkyl esters of acidic groups, including, but not limited to, carboxylic acids, phosphoric acids, phosphinic acids, sulfinic acids, sulfonic acids and boronic acids.
  • esters include formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
  • the esters can be formed with a hydroxy or carboxylic acid group of the parent compound.
  • a "pharmaceutically acceptable form” of a disclosed compound includes, but is not limited to, pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled derivatives of disclosed compounds.
  • a "pharmaceutically acceptable form” includes, but is not limited to, pharmaceutically acceptable salts, isomers, prodrugs and isotopically labeled derivatives of disclosed compounds.
  • a "pharmaceutically acceptable form” includes, but is not limited to, pharmaceutically acceptable salts, stereoisomers, prodrugs and isotopically labeled derivatives of disclosed compounds.
  • the pharmaceutically acceptable form is a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salt refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66: 1 -19.
  • Pharmaceutically acceptable salts of the compounds provided herein include those derived from suitable inorganic and organic acids and bases. Examples of
  • nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate,
  • benzenesulfonate besylate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pic
  • organic acids from which salts can be derived include, for example, acetic acid, propionic acid, gly colic acid, pyruvic acid, oxalic acid, lactic acid, trifluoracetic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • the salts can be prepared in situ during the isolation and purification of the disclosed compounds, or separately, such as by reacting the free base or free acid of a parent compound with a suitable base or acid, respectively.
  • Pharmaceutically acceptable salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci_ 4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • compositions include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines, including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
  • the pharmaceutically acceptable base addition salt can be chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
  • the pharmaceutically acceptable form is a "solvate” (e.g., a hydrate).
  • solvate refers to compounds that further include a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces.
  • the solvate can be of a disclosed compound or a pharmaceutically acceptable salt thereof. Where the solvent is water, the solvate is a "hydrate” .
  • Pharmaceutically acceptable solvates and hydrates are complexes that, for example, can include 1 to about 100, or 1 to about 10, or 1 to about 2, about 3 or about 4, solvent or water molecules. It will be understood that the term "compound” as used herein encompasses the compound and solvates of the compound, as well as mixtures thereof.
  • the pharmaceutically acceptable form is a prodrug.
  • prodrug refers to compounds that are transformed in vivo to yield a disclosed compound or a pharmaceutically acceptable form of the compound.
  • a prodrug can be inactive when administered to a subj ect, but is converted in vivo to an active compound, for example, by hydrolysis (e.g., hydrolysis in blood).
  • hydrolysis e.g., hydrolysis in blood
  • a prodrug has improved physical and/or delivery properties over the parent compound.
  • Prodrugs can increase the bioavailability of the compound when administered to a subject (e.g., by permitting enhanced absorption into the blood following oral administration) or which enhance delivery to a biological compartment of interest (e.g., the brain or lymphatic system) relative to the parent compound.
  • exemplary prodrugs include derivatives of a disclosed compound with enhanced aqueous solubility or active transport through the gut membrane, relative to the parent compound.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7- 9, 21 -24 (Elsevier, Amsterdam).
  • a discussion of prodrugs is provided in Higuchi, T., et al., "Prodrugs as Novel Delivery Systems," A. C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
  • Exemplary advantages of a prodrug can include, but are not limited to, its physical properties, such as enhanced water solubility for parenteral administration at physiological pH compared to the parent compound, or it can enhance absorption from the digestive tract, or it can enhance drug stability for long-term storage.
  • the term "pharmaceutically acceptable" excipient, carrier, or diluent refers to a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body.
  • a pharmaceutically acceptable material, composition or vehicle such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically-acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol;
  • polyols such as glycerin, sorbitol, mannitol and polyethylene glycol
  • esters such as ethyl oleate and ethyl laurate
  • agar buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
  • wetting agents such as sodium lauryl sulfate, magnesium stearate, and polyethylene oxide-polypropylene oxide copolymer as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • the term “subject” refers to any animal (e.g., a mammal), including, but not limited to humans, non-human primates, rodents, and the like, which is to be the recipient of a particular treatment.
  • the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.
  • the "low dosage” refers to at least 5% less (e.g., at least 10%, 20%, 50%, 80%, 90%, or even 95%) than the lowest standard recommended dosage of a particular compound formulated for a given route of administration for treatment of any human disease or condition.
  • the "high dosage” is meant at least 5% (e.g., at least 10%, 20%, 50%, 100%, 200%, or even 300%) more than the highest standard recommended dosage of a particular compound for treatment of any human disease or condition.
  • Compounds of the present invention are, subsequent to their preparation, preferably isolated and purified to obtain a composition containing an amount by weight equal to or greater than 95% ("substantially pure"), which is then used or formulated as described herein. In certain embodiments, the compounds of the present invention are more than 99% pure.
  • Solvates and polymorphs of the compounds of the invention are also contemplated herein.
  • Solvates of the compounds of the present invention include, for example, hydrates.
  • the invention provides novel chemical entities that are biochemically potent and physiologically active with improved pharmacokinetic and toxicological properties over 5-fluoro-3- phenyl-2-[(l S)-l-(9H-purin-6-ylamino)propyl]quinazolin-4(3H)-one (Idelalisib), shown below.
  • Idelalisib (Zydelig ® ) is an inhibitor of phosphatidylinositol 3-kinase (PI3K), approved for use in combination with rituximab for the treatment of patients with relapsed chronic lymphocytic leukemia (CLL) for whom rituximab alone would be considered appropriate therapy due to other comorbidities. Idelalisib is also indicated to treat patients with relapsed follicular B-cell non-Hodgkin lymphoma (FL) or relapsed small lymphocytic lymphoma (SLL) who have received at least two prior systemic therapies.
  • FL relapsed follicular B-cell non-Hodgkin lymphoma
  • SLL small lymphocytic lymphoma
  • the compounds disclosed herein are deuterium-substituted versions of idelalisib, where hydrogen is replaced with deuterium at strategic locations of the molecule.
  • the substitution locations are selected with the specific objective to impact pharmacokinetic and toxicological properties of the molecule.
  • Dy-Idelalisib [0051] The preferred compound, shown above, has 7 deuterium substitutions and exhibits superior safety, efficacy and tolerability profiles. The compound affords new and improved options in the treatment of blood cancers and related diseases and conditions, such as CLL. The compound may also be used to treat FL and SLL (e.g., relapsed FL or SLL, in particular after prior systemic therapies).
  • FL and SLL e.g., relapsed FL or SLL, in particular after prior systemic therapies.
  • GS-563117 was a significant metabolite in humans (exposures [AUC] were 3.3 times those of idelalisib), it was only a minor metabolite in animal species (exposures [AUC] to GS- 563117 were 0.7-3.6%, 4.4-8.9% and 16-66% those of idelalisib in rats, rabbits and dogs, respectively). Researches to date indicated that this oxidation product (formed by purine ring oxidation on carbon-8) may be responsible for the skin toxicity, reproductive toxicity and phototoxicity of Idelalisib.
  • GS-563117 has significant inhibitory activity on Ste20-Like Kinase (SLK) and
  • Lymphocyte-Oriented Kinase at clinically relevant concentrations.
  • the pharmacological activity oxidation product at carbon-8 of the purine ring is against the LOK and SLK kinases that may activate lymphocytic response and lead to skin disorder.
  • GS-563117 was clearly shown to be phototoxic to cultured cells (IC50 16-23 ⁇ g/mL; ERC max 5-7) in in vitro experiment. In tissue distribution studies, there appeared to be some retention of drug related material in pigmented tissues. Since idelalisib and GS-563117 absorbs light, distributes and is retained in pigmented skin, and the metabolite GS-563117 was shown to be phototoxic in vitro, some phototoxic skin reactions may be seen in vivo.
  • GS-563117 inhibits the catalytic activities of cytochrome P450 enzymes CYP2C9, CYP3A, CYP2C8 and CYP2C19.
  • Some patients were co-administered CYP2C8, CYP2C9 or UGT1 Al substrates, especially about 30% of patients enrolled in NHL and CLL trials were coadministered sensitive CYP2C19 substrates, including lansoprazole and omeprazole.
  • the incidence of diarrhea and rash were higher in patients taking proton pump inhibitors (PPI). These adverse events are associated with both PPI and idelalisib.
  • idelalisib The exposure to idelalisib is similar in patients taking acid-reducing agents (ARA) as compared to patients not taking these agents. Overlapping adverse events or an increased exposure to PPI (CYP2C19 substrates) may explain the increased incidence of adverse events. Higher exposures (5-12 times) for PPI have been observed in poor CYP2C19 metabolizers and dose-response has been observed with diarrhea and infections.
  • ARA acid-reducing agents
  • GS-56311 as its plasma concentration is much higher than idelalisib in human, may cause toxicities through drug-drug interaction (DDI) in patients taking other medicines.
  • the invention aims at reducing metabolite caused toxicities while increasing parent drug exposure.
  • the invention provides compounds that are orally available phosphoinositide 3-kinase (PI3K) delta inhibitors.
  • PI3K phosphoinositide 3-kinase
  • the compounds target PI 105, the delta isoform of PI3K, one of the proteins responsible for leukemia and other cell growth.
  • the invention generally relates to a compound having the structural formula of:
  • R 1; R2, R3, R4, R5, R6 and R7 is D
  • the invention generally relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound having the structural formula of:
  • each of R 1; R2, R3, R4, R5, R6 and R7 is D, or a pharmaceutically acceptable salt or ester thereof, effective to treat, prevent, or reduce one or more blood cancers, or a related disease or disorder thereof, in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier, or diluent.
  • the invention generally relates to a unit dosage form comprising the pharmaceutical composition disclosed herein.
  • the unit dosage is suitable for administration to a subject suffering from one or more blood cancers, including advanced hematologic malignancies, or a related disease or disorder thereof.
  • the invention generally relates to a method for treating, reducing, or preventing a disease or disorder.
  • the method includes: administering to a subject in need thereof a pharmaceutical composition comprising compound having the formula of:
  • each of R 1; R 2 , R 3 , R4, R5, R6 and R7 is D, or a pharmaceutically acceptable salt or ester thereof, effective to treat, prevent, or reduce one or more blood cancers, or related a related disease or disorder thereof.
  • the one or more blood cancers include diseases and conditions that may benefit from treatment using the compounds, pharmaceutical composition, unit dosage form and treatment method disclosed herein include any diseases and disorders that may be addressed through inhibition of the delta isoform of PI3K, such as ALL, FL and SLL.
  • the method of treatment includes administering to a subject in need thereof a pharmaceutical composition comprising compound having the formula of:
  • each of R 1; R 2 , R 3 , R4, R5, R6 and R7 is D, or a pharmaceutically acceptable salt or ester thereof, in combination with one or more other anti-cancer agents.
  • the one or more other anti-cancer agents may be a small molecule, a chemotherapeutic agent, a peptide, a polypeptide or protein, an antibody, an antibody-drug conjugate, an aptamer or nucleic acid molecule.
  • the one or more other anti-cancer agents are chemotherapeutic agents, chemical compounds useful in the treatment of cancer.
  • chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sutent (SU11248, Pfizer), Letrozole
  • CYTOXAN® cyclosphosphamide alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and
  • methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin;
  • nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g. , calicheamicin, especially calicheamicin gammall and calicheamicin omegall (Angew Chem. Intl. Ed. Engl. (1994) 33: 183-186); dynemicin, including dynemicin A;
  • bisphosphonates such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6- diazo-5-oxo-L-norleucine,
  • ADRIAMYCIN ® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2- pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esonibicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamniprine, thioguanine
  • TAXOTERE ® (doxetaxel; Rhone-Poulenc Rorer, Antony, France); chloranmbucil; GEMZAR ® (gemcitabine); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE ® (vinorelbine); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine
  • XELODA ® ibandronate
  • CPT-11 topoisomerase inhibitor
  • RFS 2000 difluoromethylomithine
  • retinoids such as retinoic acid
  • the one or more other anti-cancer agents are selected from monoclonal antibodies against CD20 protein, for example, rituximab.
  • Rituximab is a chimeric monoclonal antibody against CD20 protein, which is primarily found on the surface of immune system B cells.
  • Rituximab is used to treat diseases characterized by excessive numbers of B cells, overactive B cells, or dysfunctional B cells, including many lymphomas, leukemias, transplant rejection, and autoimmune disorders.
  • the compounds disclosed herein may serve as second line drugs for patients whose CLL has relapsed. These compounds may be used in combination with rituximab in patients when CLL has come back after prior cancer treatment. These compounds are particularly effective in patients who have a p53 mutation, which otherwise tends to impart a poor prognosis in CLL patients.
  • Any appropriate route of administration can be employed, for example, parenteral, intravenous, subcutaneous, intramuscular, intraventricular, intracorporeal, intraperitoneal, rectal, or oral administration. Most suitable means of administration for a particular patient will depend on the nature and severity of the disease or condition being treated or the nature of the therapy being used and on the nature of the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the compounds described herein or derivatives thereof are admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (i) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (ii) binders, as for example, carboxymethylcellulose, alignates, gelatin,
  • inert customary excipient such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid
  • binders as for example, carboxymethylcellulose, alignates, gelatin
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate
  • solution retarders as for example, paraffin
  • absorption accelerators as for example, quaternary ammonium compounds
  • wetting agents as for example, cetyl alcohol, and glycerol monostearate
  • adsorbents as for example, kaolin and bentonite
  • lubricants as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof.
  • the dosage forms may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others known in the art.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers, such as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols, and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • the composition can also benzoate, propyleneglycol, 1,3-butylenegly
  • Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis- and frafts-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • Isomeric mixtures containing any of a variety of isomer ratios may be utilized in accordance with the present invention. For example, where only two isomers are combined, mixtures containing 50:50, 60:40, 70:30, 80:20, 90: 10, 95:5, 96:4, 97:3, 98:2, 99: 1, or 100:0 isomer ratios are contemplated by the present invention. Those of ordinary skill in the art will readily appreciate that analogous ratios are contemplated for more complex isomer mixtures.
  • a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers.
  • the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic methods well known in the art, and subsequent recovery of the pure enantiomers.
  • Trifluoroacetic acid (10 mL). The reaction mixture was stirred for 1 hour, concentrated in vacuo, and partitioned between DCM and 10% potassium carbonate (aqueous layer pH -10 after extraction). The aqueous layer was extracted with additional DCM, and the combined organic layer was washed with water and brine, and dried with magnesium sulfate anhydrous. The solution was concentrated to provide compound H as an off-white solid (2.42 g, 87% yield). 1 HNMR (CDCI 3 ) is shown in FIG. 5.
  • n-BuLi 2.5 M, 23.5 mL, 58.7 mmol, 1.4 equiv
  • diisopropylamine 5.94 g, 58.7 mmol, 1.4 equiv
  • THF tetrahydrofuran
  • 6-chloro-9-(tetrahydro-2H-pyran-2-yl)-9H-purine (10 g, 41.9 mmol, 1 equiv) in THF (20 mL) solution was added drop-wise, while maintaining the temperature below -68 °C.
  • the mixture was stirred for 1 hour and then 5 mL D 2 0 was added.
  • the separated THF layer was concentrated.
  • the aqueous phase was extracted with EtOAc (100 mL x 2).
  • the EtOAc layer was combined with concentred THF and washed with brine (75 mL x 2), and dried over Na 2 S0 4 .
  • Cryopreserved hepatocytes represent a well-accepted experimental system for the evaluation of drug properties including metabolic stability, metabolite identification, drug-drug interaction potential, and hepatotoxic potential.
  • Cryopreserved human hepatocytes were obtained from IV AL LLC.
  • the donor characteristics of the hepatocyte lot (HH1009) used for the study was as follows:
  • test article was administered in vitro directly or through a solvent compatible with the test system. Studies were performed in uncoated 24-well plates.
  • Cryopreserved hepatocytes were thawed in a 37 °C water bath and placed on ice.
  • the thawed hepatocytes were recovered using Universal Cryopreserved Recovery MediumTM (UCRMTM) and centrifuged at 100 x g for 5 minutes to remove residual cryopreservants.
  • the hepatocyte pellet was re-suspended in William's E based medium HIM. Viability and cell concentration were determined based on trypan blue exclusion using a hemacytometer. The cell suspension was adjusted to 1.11 x 10 6 cells per mL and placed on ice until use.
  • the final reaction mixture for hepatocyte metabolism consisted of HIM, hepatocytes at 1 x 10 6 cells per mL and the test article or negative controls.
  • the study was designed such that a reference article (idelalisib) was used along with the test article (D 7 -idelalisib) per group.
  • the final concentration of reference article and the test article at the initiation of incubation was 2 ⁇ .
  • Each test article including reference article was prepared as 20,000 x stock in DMSO at concentration of 40 mM.
  • Each test article was mixed with reference article in equal volume to prepare a combined 10,000 x DMSO stock of 20 mM each. This DMSO stock is diluted 1000 x in HIM to prepare a 10 x dosing stock of 20 ⁇ . This dosing stock was diluted to 2 ⁇ upon addition to medium containing hepatocytes or blank medium.
  • Incubation of the hepatocytes with 2 ⁇ test article + reference article was performed in triplicates in a incubator maintained at constant temperature 37° C and humidified atmosphere of 5% CO2 and 95% balanced air for time periods of 0, 30, 60, 120, 180 and 240 minutes and with the concentrations of the hepatocytes at 1 x 10 6 cells per mL.
  • Negative controls included samples in the absence of hepatocytes (incubation media) but with 2 ⁇ test article + reference article only at the same six time points. These controls were conducted under identical conditions.
  • Total reaction volume was 500 ⁇ . (0.450 mL hepatocytes suspension + 0.050 mL incubation buffer media with lOx test compound or positive control). All samples except negative control were run in triplicates.
  • Test article concentrations One concentration (2.0 ⁇ ) of the test article+ reference article was used in all incubations.
  • Positive Controls The reference article at 2 ⁇ in each group served as positive control.
  • Termination After incubation, the reaction was terminated with as described previously. The total mixture after termination was stored frozen for LCMS analysis.
  • FIG. 8 shows the percentage of compounds remaining vs. incubation time. After 4 hour, the concentration of D 7 -idelalisib is approximately equal to 220% of idelalisib. The result showed D 7 - idelalisib has longer half-life and AUC. This substantial difference indicates superior DMPK property of Dy-idelalisib.
  • the oxidation product (formed by purine ring oxidation on carbon-8) may be responsible for the skin toxicity, reproductive toxicity and phototoxicity of Idelalisib. Selectively modify the molecular structure to reduce the formation of the metabolite may lead to minimize toxic side effects in human application.
  • the samples generated from the procedure described herein (para. [0086]-[0097]) were analyzed to compare the formation of the metabolite.
  • FIG. 9 compares idelalisib vs. D 7 -idelalisib in the formation of oxidative metabolites formed by purine ring oxidation on carbon-8. The results supported that D 7 -idelalisib slowed the metabolism that may cause toxicity in human disease treatment.
  • the PIP3 product was detected by displacement of biotin-PIP3 from an energy transfer complex consisting of Europium labeled anti- GST monoclonal antibody, a GST-tagged pleckstrin homology (PH) domain, biotinylated PIP3 and Streptavidin-Allophycocyanin (APC). Excitation of Europium in the complex resulted in an energy transfer to the APC and a fluorescent emission at 665 nm.
  • the PIP3 product formed by PI 3- Kinase(h) activity displaced biotin-PIP3 from the complex resulting in a loss of energy transfer and thus a decrease in signal.
  • Idelalisib and D 7 -idelalisib were received as a 2 mg/mL stock in DMSO. Both compounds were tested against 1 PI3K isoform. These compounds were tested in 10-dose IC50 mode starting at a concentration of 2 ⁇ . Control compound, PI-103, was tested in 10-dose IC50 with 3-fold serial dilution starting at 10 ⁇ . Reactions were carried out at 10 ⁇ ATP. HTRF assay format was used for POKs. Curve fits were performed where the enzyme activities at the highest concentration of compounds were less than 65%.

Landscapes

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

Abstract

L'invention concerne de nouveaux composés chimiques utiles pour traiter diverses leucémies, ou une maladie ou un trouble associé à une leucémie, ainsi qu'une composition pharmaceutique contenant ces composés, et des procédés de préparation et d'utilisation de ceux-ci.
PCT/US2016/058838 2015-11-03 2016-10-26 Composés deutérés pour le traitement de leucémies, compositions et procédés associés WO2017079003A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201680060582.XA CN108137592B (zh) 2015-11-03 2016-10-26 用于治疗血癌的氘代化合物以及其组合物和方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562249991P 2015-11-03 2015-11-03
US62/249,991 2015-11-03

Publications (1)

Publication Number Publication Date
WO2017079003A1 true WO2017079003A1 (fr) 2017-05-11

Family

ID=58662617

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/058838 WO2017079003A1 (fr) 2015-11-03 2016-10-26 Composés deutérés pour le traitement de leucémies, compositions et procédés associés

Country Status (2)

Country Link
CN (1) CN108137592B (fr)
WO (1) WO2017079003A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011028922A1 (fr) * 2009-09-02 2011-03-10 Concert Pharmaceuticals, Inc. Dérivés de xanthine substitués
WO2014194254A1 (fr) * 2013-05-30 2014-12-04 Infinity Pharmaceuticals, Inc. Traitement anticancer au moyen de modulateurs isoformes de la kinase p13
WO2015113521A1 (fr) * 2014-01-30 2015-08-06 苏州泽璟生物制药有限公司 Composé de quinazolinone deutéré et composition pharmaceutique le comprenant

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ744567A (en) * 2012-11-01 2020-03-27 Infinity Pharmaceuticals Inc Treatment of cancers using pi3 kinase isoform modulators
WO2015179772A1 (fr) * 2014-05-23 2015-11-26 Concert Pharmaceuticals, Inc. Composés phénylquinazolinone et phénylisoquinolinone deutérés

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011028922A1 (fr) * 2009-09-02 2011-03-10 Concert Pharmaceuticals, Inc. Dérivés de xanthine substitués
WO2014194254A1 (fr) * 2013-05-30 2014-12-04 Infinity Pharmaceuticals, Inc. Traitement anticancer au moyen de modulateurs isoformes de la kinase p13
WO2015113521A1 (fr) * 2014-01-30 2015-08-06 苏州泽璟生物制药有限公司 Composé de quinazolinone deutéré et composition pharmaceutique le comprenant

Also Published As

Publication number Publication date
CN108137592A (zh) 2018-06-08
CN108137592B (zh) 2021-03-05

Similar Documents

Publication Publication Date Title
EP2797919B1 (fr) Pyridophthalazinones tétra ou pentacycliques fusionnés à utiliser en tant qu'inhibiteurs de parp
EP3280715B1 (fr) Nouvelles imidazo[1,5-a]pyridines substituées en 5ème ou 8ème position en tant qu'indoleamine et/ou tryptophane 2,3-dioxygénases
CN107873032A (zh) 苯并氧氮杂*噁唑烷酮化合物及其使用方法
US11248016B2 (en) Glycolipids and pharmaceutical compositions thereof for use in therapy
WO2011135520A1 (fr) Pipérazinotriazines comme inhibiteurs de pi3k pour une utilisation dans le traitement de troubles antiprolifératifs
US10561658B2 (en) Thieno- and pyrrolopyrimidine analogues as anticancer agents and methods of use thereof
WO2017087207A1 (fr) Composés deutérés pour le traitement de maladies hématologiques malignes, inflammatoires et auto-immunes
WO2017079003A1 (fr) Composés deutérés pour le traitement de leucémies, compositions et procédés associés
US20230382920A1 (en) SHMT Inhibitors
US20180055915A1 (en) Compositions and methods for treating prostate cancer
US20240099992A1 (en) Compositions and methods for treating cancer
EP3700521B1 (fr) Utilisation d'un compose inhibiteur d'adam9 et d'une composition comprenant un compose inhibiteur d'adam9
WO2017077307A1 (fr) Thérapie et composition pharmaceutique
CN106573923B (zh) 苯氨基嘧啶衍生化合物,用于生产其的方法,所述化合物用于治疗癌症的用途和治疗方法
EP3710003A1 (fr) Composés, compositions et méthodes d'inhibition sélective des beta-glucuronidases et d'atténuation des effets secondaires associés à une diarrhée provoquée par un traitement médicamenteux
WO2020181199A1 (fr) COMPOSÉS, COMPOSITIONS ET PROCÉDÉS D'INHIBITION SÉLECTIVE DE β-GLUCURONIDASES ET DE SOULAGEMENT D'EFFETS SECONDAIRES ASSOCIÉS À LA DIARRHÉE INDUITE PAR UN TRAITEMENT MÉDICAMENTEUX
WO2024114467A1 (fr) Inhibiteurs de shp2, compositions et procédés associés

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16862722

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16862722

Country of ref document: EP

Kind code of ref document: A1