US20160046579A1 - Therapeutic compounds and compositions - Google Patents

Therapeutic compounds and compositions Download PDF

Info

Publication number
US20160046579A1
US20160046579A1 US14/776,635 US201414776635A US2016046579A1 US 20160046579 A1 US20160046579 A1 US 20160046579A1 US 201414776635 A US201414776635 A US 201414776635A US 2016046579 A1 US2016046579 A1 US 2016046579A1
Authority
US
United States
Prior art keywords
compound
alkyl
optionally substituted
aryl
heteroaryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/776,635
Other languages
English (en)
Inventor
Giovanni Cianchetta
Janeta Popovici-Muller
Robert Zahler
Sheldon Cao
Xiaolei Wang
Zhixiong Ye
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agios Pharmaceuticals Inc
Original Assignee
Agios 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51535825&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20160046579(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Agios Pharmaceuticals Inc filed Critical Agios Pharmaceuticals Inc
Publication of US20160046579A1 publication Critical patent/US20160046579A1/en
Assigned to AGIOS PHARMACEUTICALS, INC reassignment AGIOS PHARMACEUTICALS, INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZAHLER, ROBERT, CIANCHETTA, GIOVANNI, POPOVICI-MULLER, JANETA
Assigned to VIVA BIOTECH (SHANGHAI) LTD. reassignment VIVA BIOTECH (SHANGHAI) LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CAO, SHELDON, WANG, XIAOLEI, YE, ZHIXIONG
Assigned to AGIOS PHARMACEUTICALS, INC reassignment AGIOS PHARMACEUTICALS, INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VIVA BIOTECH (SHANGHAI) LTD.
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/48Oxygen atoms attached in position 4 having an acyclic carbon atom attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/52Oxygen atoms attached in position 4 having an aryl radical as the second substituent in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom

Definitions

  • PTD Pyruvate kinase deficiency
  • Human erythrocytes are unique in that they anucleate when mature. Immature erythrocytes have nuclei but during early erythropoiesis prior to becoming circulating reticulocytes they extrude nuclei as well as other organelles such as mitochondria, endoplasmic reticulum, and golgi apparatusus, in order to make room for oxygen-carrying hemoglobin. As a result of lacking mitochondria, mature red blood cells do not utilize any of the oxygen they transport to economically synthesize adenosine triphosphate (ATP) as other normal differentiated cells do.
  • ATP adenosine triphosphate
  • red blood cells depend entirely on anaerobic glycolysis to cycle nicotinamide adenine dinucleotide (NAD + ) and to make ATP, an essential energy source largely used to drive ATPase-dependent K + /Na + and Ca 2+ pumps, in order to maintain cell membrane integrity and pliability as they navigate through blood vessels.
  • NAD + nicotinamide adenine dinucleotide
  • ATP ase-dependent K + /Na + and Ca 2+ pumps
  • Pyruvate kinase catalyzes the transfer of a phosphoryl group from phosphoenolpyruvate (PEP) to ADP, yielding one molecule of pyruvate and one molecule of ATP.
  • PEP phosphoenolpyruvate
  • the enzyme has an absolute requirement for Mg 2+ and K + cations to drive catalysis.
  • PK functions as the last critical step in glycolysis because it is an essentially irreversible reaction under physiological conditions.
  • pyruvate kinase is also an important cellular metabolism regulator.
  • pyruvate kinase is tightly controlled at both gene expression and enzymatic allostere levels. In mammals, fully activated pyruvate kinase exists as a tetrameric enzyme.
  • isozymes M1, M2, L and R
  • Erythrocyte-specific isozyme PKR is expressed from the PKLR gene (“L gene”) located on chromosome 1q21.
  • PKLR consists of 12 exons with exon 1 is erythroid-specific whereas exon 2 is liver-specific.
  • the two other mammalian isozymes PKM1 and PKM2 are produced from the PKM gene (“M gene”) by alternative splicing events controlled by hnRNP proteins.
  • M gene the PKM gene
  • the PKM2 isozyme is expressed in fetal tissues and in adult proliferating cells such as cancer cells. Both PKR and PKM2 are in fact expressed in proerythroblasts. However, upon erythroid differentiation and maturation, PKM2 gradually is decreased in expression and progressively replaced by PKR in mature erythrocytes.
  • hereditary PKR deficiency disorder manifests as non-spherocytic hemolytic anemia.
  • the clinical severity of this disorder range from no observable symptoms in fully-compensated hemolysis to potentially fatal severe anemia requiring chronic transfusions and/or splenectomy at early development or during physiological stress or serious infections.
  • Most of the most severe cases while extremely rare population-wise with estimated prevalence of 51 per million (Beutler, E. Blood 2000, 95 (11), 3585-8), there is no disease-modifying treatment available for these patients other than palliative care (Tavazzi, D. et al., Pediatr Ann 2008, 37 (5), 303-10).
  • HNSHA hereditary non-spherocytic haemolytic anemia
  • Cancer cells rely primarily on glycolysis to generate cellular energy and biochemical intermediates for biosynthesis of lipids and nucleotides, while the majority of “normal” cells in adult tissues utilize aerobic respiration. This fundamental difference in cellular metabolism between cancer cells and normal cells, termed the Warburg Effect, has been exploited for diagnostic purposes, but has not yet been exploited for therapeutic benefit.
  • PK Pyruvate kinase
  • L and R isoforms are expressed in liver and red blood cells
  • M1 isoform is expressed in most adult tissues
  • M2 isoform is a splice variant of M1 expressed during embryonic development. All tumor cells exclusively express the embryonic M2 isoform.
  • M2 is a low-activity enzyme that relies on allosteric activation by the upstream glycolytic intermediate, fructose-1,6-bisphosphate (FBP), whereas M1 is a constitutively active enzyme.
  • FBP fructose-1,6-bisphosphate
  • PKM2 is also expressed in adipose tissue and activated T-cells. Phosphotyrosine peptide binding to PKM2 leads to a dissociation of FBP from PKM2 and conformational changes of PKM2 from an active, tetrameric form to an inactive form. Compounds that bind to PKM2 and lock the enzyme in the active confirmation will lead to the loss of allosteric control of PKM2 needed for shunting biochemical intermediates from glycolysis into biosynthesis of nucleotides and lipids.
  • the activation of PKM2 can inhibit the growth and proliferation of cancer cells, activated immune cells, and fat cells. Activation of PKM2 may therefore be effective in the treatment of cancer, obesity, diabetes, autoimmune conditions, and proliferation-dependent diseases, e.g., benign prostatic hyperplasia (BPH).
  • BPH benign prostatic hyperplasia
  • Described herein are compounds that activate pyruvate kinase and pharmaceutically acceptable salts, solvates, and hydrates thereof, for example, compounds that activate PKR and/or PKM2.
  • compositions comprising a compound provided herewith and the use of such compositions in methods of treating diseases and conditions that are related to pyruvate kinase function, e.g., PKR function, and/or PKM2 function (including, e.g., cancer, diabetes, obesity, autoimmune disorders, and benign prostatic hyperplasia (BPH)).
  • diseases and conditions that are related to pyruvate kinase function, e.g., PKR function, and/or PKM2 function (including, e.g., cancer, diabetes, obesity, autoimmune disorders, and benign prostatic hyperplasia (BPH)).
  • A is aryl or heteroaryl, wherein the aryl or heteroaryl is optionally substituted, and the aryl or heteroaryl is optionally fused to an optionally substituted carbocyclyl or an optionally substituted heterocyclyl;
  • X is selected from —NH—S(O) 2 —, —S(O) 2 —NH—, —NH—S(O) 2 —CH 2 —, —CH 2 —S(O)—NH—, —NH—S(O)—CH 2 —, —NH—S(O)—, —S(O)—NH—, or —CH 2 —S(O) 2 —NH—;
  • Y is C(H) or N; provided that no more than two Y groups are N;
  • R 1a is hydroxyl, —CH 2 OH, —CHO, —CO 2 H, —N(R 10a ) 2 , —CO 2 —C 1-6 alkyl, —OP( ⁇ O)(OH) 2 , or —OCO 2 —CH 2 —OP( ⁇ O)(OH) 2 ;
  • R 1b is C 1-8 alkyl optionally substituted with one to four R 5 groups; C 1-8 alkenyl optionally substituted with one to four R 5 groups; cycloalkyl; heterocycle; aryl; heteroaryl; cycloalkylalkyl; cycloalkylalkenyl; heterocyclylalkyl; heterocyclylalkenyl; aralkyl; aralkenyl; heteroaralkyl; heteroaralkenyl; or —OH, with the proviso that when R 1a is OH, R 1b is not OH; wherein each cycloalkyl, heterocycle, aryl, heteroaryl, cycloalkylalkyl, cycloalkylalkenyl, heterocyclylalkyl, heterocyclylalkenyl, aralkyl, aralkenyl, heteroaralkyl, or heteroaralkenyl is optionally substituted;
  • each R 2 is independently selected from halo, alkyl, CN, OH, and alkoxy, wherein said alkyl or alkoxy is optionally substituted with one to four R 5 groups; or
  • R 2 groups are taken together with the ring atoms they are attached to form a 5- or 6-membered carbocyclic, aryl, heterocyclic or heteroaryl ring;
  • each R 4 is independently selected from halo, alkyl, alkoxy, haloalkyl, haloalkoxy and hydroxyl;
  • each R 5 is independently selected from halo, OH, C 1-6 alkoxy, CN, NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl) 2 ;
  • each R 10a is independently selected from hydrogen or C 1-6 alkyl
  • n 0, 1, 2 or 3;
  • n 0, 1 or 2; provided that a compound of Formula (I) is not the following:
  • a method for treating or preventing comprising administering a compound provided herein, a pharmaceutically acceptable salt, solvate or hydrate thereof, or pharmaceutical composition thereof.
  • a method for increasing lifetime of the red blood cells (RBCs) in need thereof comprising contacting blood with an effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a composition comprising a compound disclosed herein or a salt, solvate or hydrate thereof and a carrier; or (3) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for regulating 2,3-diphosphoglycerate levels in blood in need thereof comprising contacting blood with an effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a composition comprising a compound disclosed herein or a salt, solvate or hydrate thereof and a carrier; or (3) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating hereditary non-spherocytic haemolytic anemia comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating sickle cell anemia comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • hemolytic anemia e.g., chronic hemolytic anemia caused by phosphoglycerate kinase deficiency, Blood Cells Mol Dis, 2011; 46(3):206
  • administering comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating diseases or conditions that are associated with increased 2,3-diphosphoglycerate levels comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • thalassemia e.g., beta-thalassemia
  • hereditary spherocytosis hereditary elliptocytosis
  • abetalipoproteinemia or Bassen-Kornzweig syndrome
  • paroxysmal nocturnal hemoglobinuria acquired hemolytic anemia (e.g., congenital anemias (e.g., enzymopathies))
  • anemia of chronic diseases comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating diseases or conditions that are associated with increased 2,3-diphosphoglycerate levels comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • PKR mutants having lower activities compared to the wild type, thus are useful for methods of the present invention.
  • Such mutations in PKR can affect enzyme activity (catalytic efficiency), regulatory properties (modulation by fructose bisphosphate (FBP)/ATP), and/or thermostability of the enzyme. Examples of such mutations are described in Valentini et al, JBC 2002.
  • Some examples of the mutants that are activated by the compounds described herein include G332S, G364D, T384M, G37E, R479H, R479K, R486W, R532W, R510Q, and R490W.
  • compounds described herein affect the activities of PKR mutants by activating FBP non-responsive PKR mutants, restoring thermostability to mutants with decreased stability, or restoring catalytic efficiency to impaired mutants.
  • the activating activity of the present compounds against PKR mutants may be tested following a method described in Examples 2-5.
  • Compounds described herein are also activators of wild type PKR.
  • a compound, composition or pharmaceutical composition described herein is added directly to whole blood or packed cells extracorporeally or be provided to the subject (e.g., the patient) directly (e.g., by i.p., i.v., i.m., oral, inhalation (aerosolized delivery), transdermal, sublingual and other delivery routes).
  • compounds described herein increase the lifetime of the RBCs, thus counteract aging of stored blood, by impacting the rate of release of 2,3-DPG from the blood.
  • a decrease in the level of 2, 3-DPG concentration induces a leftward shift of the oxygen-hemoglobin dissociation curve and shifts the allosteric equilibribrium to the R, or oxygenated state, thus producing a therapeutic inhibition of the intracellular polymerization that underlies sickling by increasing oxygen affinity due to the 2,3-DPG depletion, thereby stabilizing the more soluble oxy-hemoglobin.
  • compounds and pharmaceutical compositions described herein are useful as antisickling agents.
  • a compound, composition or pharmaceutical composition described herein is added directly to whole blood or packed cells extracorporeally or be provided to the subject (e.g., the patient) directly (e.g., by i.p., i.v., i.m., oral, inhalation (aerosolized delivery), transdermal, sublingual and other delivery routes).
  • a method of increasing the level of PKM2 activity and/or glycolysis in a patient in need thereof comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby increasing the level of PKM2 activity and/or glycolysis in the patient.
  • a compound or a composition described herein is used to maintain PKM2 in its active conformation or activate pyruvate kinase activity in proliferating cells as a means to divert glucose metabolites into catabolic rather than anabolic processes in the patient.
  • a method of inhibiting cell proliferation in a patient in need thereof comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby inhibiting cell proliferation in the patient.
  • this method can inhibit growth of a transformed cell, more specifically a cancer cell.
  • the method generally inhibits growth of a PKM2-dependent cell that undergoes aerobic glycolysis.
  • a method of treating a patient suffering from or susceptible to a disease or disorder associated with reduced PKM2 activity or reduced glycolysis in a patient in need thereof comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby treating, preventing or ameliorating the disease or disorder in the patient.
  • the compound described herein is provided in a pharmaceutical composition.
  • the method includes the step of identifying or selecting a patient who would benefit from activation of PKM2 prior to treatment. Identifying or selecting such a patient can be on the basis of the level of PKM2 activity in a cell of the patient.
  • the selected patient is suffering from or susceptible to unwanted cell growth or proliferation, e.g., cancer, obesity, diabetes, atherosclerosis, restenosis, and autoimmune diseases.
  • the selected patient is suffering from a cancer associated with PKM2 function.
  • the compound described herein is administered at a dosage and frequency sufficient to increase lactate production or oxidative phosphorylation.
  • halo or halogen refers to any radical of fluorine, chlorine, bromine or iodine.
  • alkyl refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms.
  • C 1 -C 12 alkyl indicates that the group may have from 1 to 12 (inclusive) carbon atoms in it.
  • the term “alkyl” refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing 1 to 6 carbon atoms.
  • the term “alkyl” refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing 1 to 4 carbon atoms.
  • haloalkyl refers to an alkyl in which one or more hydrogen atoms are replaced by halo, and includes alkyl moieties in which all hydrogens have been replaced by halo (e.g., perfluoroalkyl).
  • haloalkoxy refers to an alkoxy in which one or more hydrogen atoms are replaced by halo.
  • alkenyl refers to a monovalent straight or branched hydrocarbon chain containing 2-12 carbon atoms and having one or more double bonds.
  • alkenyl groups include, but are not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups.
  • One of the double bond carbons may optionally be the point of attachment of the alkenyl substituent.
  • alkenyl refers to a monovalent straight or branched hydrocarbon chain containing 2-6 carbon atoms and having one or more double bonds.
  • alkenyl refers to a monovalent straight or branched hydrocarbon chain containing 2-4 carbon atoms and having one or more double bonds.
  • alkoxy refers to an —O-alkyl radical.
  • aryl refers to a monocyclic, bicyclic, or tricyclic aromatic hydrocarbon ring system.
  • aryl moieties include, but are not limited to, phenyl, naphthyl, and anthracenyl.
  • arylalkyl or “aralkyl” refer to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group.
  • Aralkyl includes groups in which more than one hydrogen atom has been replaced by an aryl group. Examples of “arylalkyl” or “aralkyl” include benzyl, 2-phenylethyl, 3-phenylpropyl, 9-fluorenyl, benzhydryl, and trityl groups.
  • Carbocyclyl refers to a non-aromatic, monocyclic, bicyclic, or tricyclic hydrocarbon ring system.
  • Carbocyclyl groups include fully saturated ring systems (e.g., cycloalkyls), and partially saturated ring systems.
  • cycloalkyl or “carbocyclyl” refers to saturated or unsaturated cyclic, bicyclic, tricyclic, or polycyclic non-aromatic hydrocarbon groups having 3 to 12 carbons.
  • cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclohexyl, methylcyclohexyl, adamantyl, and norbornyl.
  • heteroaryl refers to a fully aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms selected independently from N, O, or S if monocyclic, bicyclic, or tricyclic, respectively).
  • heterocyclyl refers to a saturated or unsaturated, 3-10 membered non-aromatic monocyclic, 8-12 membered non-aromatic bicyclic, or 11-14 membered non-aromatic tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively).
  • the heteroatom may optionally be the point of attachment of the heterocyclyl substituent.
  • heterocyclyl examples include, but are not limited to, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, pyrrolinyl, pyrimidinyl, and pyrrolidinyl.
  • Bicyclic and tricyclic ring systems containing one or more heteroatoms and both aromatic and non-aromatic rings are considered to be heterocyclyl groups according to the present definition.
  • Such bicyclic or tricyclic ring systems are considered to be an aryl or a heteroaryl fused to a carbocyclyl or heterocyclyl, where the ring bound to the rest of the molecule is required to be aromatic.
  • heteroarylalkyl and “heteroaralkyl”, as used herein, refers to an alkyl group substituted with a heteroaryl group.
  • heterocyclylalkyl refers to an alkyl group substituted with a heterocyclyl group.
  • All ring systems i.e, aryl, heteroaryl, carbocyclyl, cycloalkyl, heterocyclyl, etc.
  • ring system portions of groups are optionally substituted at one or more substitutable carbon atoms with substituents independently selected from: halo, —C ⁇ N, C 1 -C 4 alkyl, ⁇ O, C 3-7 cycloalkyl, C 1-6 alkyl, —OH, —O—(C 1-6 alkyl), —SO 2 —(C 1-6 alkyl), —(C 1-4 alkyl)-N(R ⁇ )(R ⁇ ), —N(R ⁇ )(R ⁇ ), —O—(C 1-4 alkyl)-N(R ⁇ )(R ⁇ ), —C(O)—N(R ⁇ )(R ⁇ ), —(C 1-4 alkyl)-N(R ⁇ )(R ⁇ ), —(O)—
  • All heterocyclyl ring systems (and any heterocyclyl substituents on any ring system) are optionally substituted on one or more any substitutable nitrogen atom with —C 1-4 alkyl, or fluoro-substituted C 1-4 alkyl.
  • substituted refers to the replacement of a hydrogen atom by another group.
  • oxo refers to an oxygen atom, which forms a carbonyl when attached to carbon, an N-oxide when attached to nitrogen, and a sulfoxide or sulfone when attached to sulfur.
  • selective in association with a PKM2 activator is meant at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, or 10-fold greater activation of PKM2 than PKM1.
  • activator of pyruvate kinase R means an agent that (measurably) increases the activity of wild type pyruvate kinase R (wtPKR) or causes wild type pyruvate kinase R (wt PKR) activity to increase to a level that is greater than wt PKR's basal levels of activity or an agent that (measurably) increases the activity of a mutant pyruvate kinase R (mPKR) or causes mutant pyruvate kinase R (mPKR) activity to increase to a level that is greater than that mutant PKR's basal levels of activity, for examples, to a level that is 20%, 40%, 50%, 60%, 70%, 80%, 90% or 100% of the activity of wild type PKR.
  • wtPKR wild type pyruvate kinase R
  • mPKR mutant pyruvate kinase R
  • mPKR mutant pyruvate
  • activator of pyruvate kinase M2 means an agent that (measurably) increases the activity of PKM2 or causes PKM2 activity to increase to a level that is greater than PKM2's basal levels of activity.
  • the activator may mimic the effect caused by a natural ligand (e.g., FBP).
  • the activator effect caused by a compound provided herein may be to the same, or to a greater, or to a lesser extent than the activating effect caused by a natural ligand, but the same type of effect is caused.
  • a compound provided herein can be evaluated to determine if it is an activator by measuring either directly or indirectly the activity of the pyruvate kinase when subjected to said compound.
  • the activity of PKM2 can be measured, for example, by monitoring the concentration of a substrate such as ATP or NADH.
  • Me, Et, Ph, Tf, Nf, Ts, Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl and methanesulfonyl, respectively.
  • a more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the Journal of Organic Chemistry ; this list is typically presented in a table entitled Standard List of Abbreviations. The abbreviations contained in said list, and all abbreviations utilized by organic chemists of ordinary skill in the art are hereby incorporated by reference.
  • a compound of Formula (I) or a pharmaceutically acceptable salt, solvate or hydrate thereof as described above in the Summary of the Invention e.g, useful for activating wild type PKR and/or various mutant PKRs such as those mutants described herein, and/or useful for selectively activating PKM2.
  • A is aryl or heteroaryl, wherein the aryl or heteroaryl is optionally substituted, and the aryl or heteroaryl is optionally fused to an optionally substituted carbocyclyl or an optionally substituted heterocyclyl;
  • X is selected from —NH—S(O) 2 —, —S(O) 2 —NH—, —NH—S(O) 2 —CH 2 —, —CH 2 —S(O)—NH—, —NH—S(O)—CH 2 —, —NH—S(O)—, —S(O)—NH—, or —CH 2 —S(O) 2 —NH—;
  • Y is C(H) or N; provided that no more than two Y groups are N;
  • R 1a is hydroxyl, —CH 2 OH, —CHO, —CO 2 H, —N(R 10a ) 2 , —CO 2 —C 1-6 alkyl, —OP( ⁇ O)(OH) 2 , or —OCO 2 —CH 2 —OP( ⁇ O)(OH) 2 ;
  • R 1b is C 1-8 alkyl optionally substituted with one to four R 5 groups; C 1-8 alkenyl optionally substituted with one to four R 5 groups; cycloalkyl; heterocycle; aryl; heteroaryl; cycloalkylalkyl; cycloalkylalkenyl; heterocyclylalkyl; heterocyclylalkenyl; aralkyl; aralkenyl; heteroaralkyl; heteroaralkenyl; or —OH, with the proviso that when R 1a is OH, R 1b is not OH; wherein each cycloalkyl, heterocycle, aryl, heteroaryl, cycloalkylalkyl, cycloalkylalkenyl, heterocyclylalkyl, heterocyclylalkenyl, aralkyl, aralkenyl, heteroaralkyl, or heteroaralkenyl is optionally substituted;
  • each R 2 is independently selected from halo, alkyl, CN, OH, and alkoxy, wherein said alkyl or alkoxy is optionally substituted with one to four R 5 groups; or
  • R 2 groups are taken together with the ring atoms they are attached to form a 5- or 6-membered carbocyclic, aryl, heterocyclic or heteroaryl ring;
  • each R 4 is independently selected from halo, alkyl, alkoxy, haloalkyl, haloalkoxy and hydroxyl;
  • each R 5 is independently selected from halo, OH, C 1-6 alkoxy, CN, NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl) 2 ;
  • each R 10a is independently selected from hydrogen or C 1-6 alkyl
  • n 0, 1, 2 or 3;
  • n 0, 1 or 2; provided that a compound of Formula (I) is not the following:
  • A is aryl or heteroaryl, wherein the aryl or heteroaryl is optionally substituted, and the aryl or heteroaryl is optionally fused to an optionally substituted carbocyclyl or an optionally substituted heterocyclyl;
  • X is selected from —NH—S(O) 2 —, —NH—S(O) 2 —CH 2 —, —CH 2 —S(O)—NH— or —CH 2 —S(O) 2 —NH—;
  • Y is C(H) or N; provided that no more than two Y groups are N;
  • R 1a is hydroxyl, —CH 2 OH, —CHO, —CO 2 H or —CO 2 —C 1-6 alkyl;
  • R 1b is C 1-8 alkyl optionally substituted with one to four R 5 groups; C 1-8 alkenyl optionally substituted with one to four R 5 groups; cycloalkyl; heterocycle; aryl; heteroaryl; cycloalkylalkyl; cycloalkylalkenyl; heterocyclylalkyl; heterocyclylalkenyl; aralkyl; aralkenyl; heteroaralkyl; heteroaralkenyl; or —OH, with the proviso that when R 1a is OH, R 1b is not OH; wherein each cycloalkyl, heterocycle, aryl, heteroaryl, cycloalkylalkyl, cycloalkylalkenyl, heterocyclylalkyl, heterocyclylalkenyl, aralkyl, aralkenyl, heteroaralkyl, or heteroaralkenyl is optionally substituted;
  • each R 2 is independently selected from halo, alkyl, CN, OH, and alkoxy, wherein said alkyl or alkoxy is optionally substituted with one to four R 5 groups; or
  • R 2 groups are taken together with the ring atoms they are attached to form a 5- or 6-membered carbocyclic, aryl, heterocyclic or heteroaryl ring;
  • each R 4 is independently selected from halo, alkyl, alkoxy, haloalkyl, haloalkoxy and hydroxyl;
  • each R 5 is independently selected from halo, OH, C 1-6 alkoxy, CN, NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl) 2 ;
  • n 0, 1, 2 or 3;
  • n 0, 1 or 2; provided that a compound of Formula (I) is not the following:
  • R 4 is hydroxyl.
  • n is 0. In certain aspects of formula (I) or (Ia), n is 1.
  • R 2 is C 1-6 alkyl (e.g., methyl).
  • R 2 is C 1-6 alkoxy (e.g., methoxy).
  • R 2 is halo (e.g., fluoro or chloro).
  • R 2 is C 4 haloalkoxy (e.g., trifluoromethoxy or difluoromethoxy).
  • R 2 is cyano.
  • n is 2.
  • each R 2 moiety is halo (e.g., fluoro).
  • A is an optionally substituted monocyclic aryl.
  • A is an optionally substituted phenyl (e.g., 2,3-dichlorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 3-cyanophenyl, 4-cyanophenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 3-trifluoromethoxyphenyl, 4-trifluoromethoxyphenyl, 2,3-diamino-4-fluorophenyl).
  • phenyl e.g., 2,3-dichlorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 3-cyanophenyl, 4-cyanophenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 3-trifluoromethoxyphenyl, 4-
  • A is an optionally substituted bicyclic aryl.
  • A is an optionally substituted naphthyl (e.g., unsubstituted naphthyl).
  • A is an optionally substituted monocyclic heteroaryl.
  • A is an optionally substituted pyridyl (e.g., an optionally substituted 3-pyridyl or optionally substituted 2-pyridyl).
  • A is unsubstituted 3-pyridyl.
  • A is unsubstituted 2-pyridyl.
  • A is an optionally substituted bicyclic heteroaryl.
  • A is an optionally substituted quinolin-8-yl (e.g., unsubstituted quinolin-8-yl).
  • A is substituted quinolin-8-yl (e.g., 2-fluoroquinolin-8-yl, 3-fluoroquinolin-8-yl, 5-fluoroquinolin-8-yl or 6-fluoroquinolin-8-yl).
  • A is an optionally substituted quinolin-3-yl (e.g., unsubstituted quinolin-3-yl).
  • A is an optionally substituted quinolin-5-yl (e.g., unsubstituted quinolin-5-yl or 2-fluoroquinolin-5-yl). In another more specific aspect, A is an optionally substituted isoquinolin-5-yl (e.g., unsubstituted isoquinolin-5-yl). In another more specific aspect, A is an optionally substituted quinolin-5-yl (e.g., unsubstituted quinolin-5-yl). In another more specific aspect, A is substituted quinolin-5-yl (e.g., 2-fluoroquinolin-5-yl).
  • A is an optionally substituted benzo[1,2,5]oxadiazole (e.g., unsubstituted benzo[1,2,5]oxadiazole).
  • A is an optionally substituted quinoxalin-5-yl (e.g., unsubstituted quinoxalin-5-yl or 8-hydroxyquinoxalin-5-yl).
  • A is substituted quinoxalin-5-yl (e.g., 8-fluoroquinoxalin-5-yl or 8-hydroxyquinoxalin-5-yl).
  • A is an optionally substituted chromanyl (e.g., chroman-8-yl).
  • A is an optionally substituted 2,3-dihydrobenzo[b][1,4]dioxinyl (e.g., unsubstituted 2,3-dihydrobenzo[b][1,4]dioxin-5-yl).
  • A is an optionally substituted benzo[d]thiazol-4-yl (e.g., unsubstituted benzo[d]thiazol-4-yl, 6-fluorobenzo[d]thiazol-4-yl, 7-fluorobenzo[d]thiazol-4-yl, 2-methylbenzo[d]thiazol-4-yl or 2-amino-6-fluorobenzo[d]thiazol-4-yl).
  • A is an optionally substituted benzofuranyl (e.g., benzofuran-7-yl).
  • A is an optionally substituted benzo[1,2,5]thiadiazol-5-yl (e.g., unsubstituted benzo[1,2,5]thiadiazol-5-yl).
  • A is an optionally substituted benzo[1,2,5]thiadiazol-4-yl (e.g., unsubstituted benzo[1,2,5]thiadiazol-4-yl).
  • A is an optionally substituted benzo[c]thiazolyl (e.g., unsubstituted benzo[c]thiazol-4-yl).
  • A is an optionally substituted 1,2,3,4-tetrahydroquinolin-8-yl (e.g., unsubstituted 1,2,3,4-tetrahydroquinolin-8-yl).
  • A is an optionally substituted 1H-indol-2(7aH)-on-5-yl (e.g., 1-methyl-1H-indol-2(7aH)-on-5-yl).
  • A is an optionally substituted thieno[3,2-b]pyridin-3-yl (e.g., unsubstituted thieno[3,2-b]pyridin-3-yl).
  • A is an optionally substituted benzo[1,3]dioxol-5-yl (e.g., unsubstituted benzo[1,3]dioxol-5-yl or 2,2-difluorobenzo[1,3]dioxol-5-yl).
  • A is an optionally substituted benzo[d]thiazol-7-yl (e.g., unsubstituted benzo[d]thiazol-7-yl or 6-methylbenzo[d]thiazol-7-yl or 6-fluorobenzo[d]thiazol-7-yl).
  • A is an optionally substituted cinnolin-8-yl (e.g., unsubstituted cinnolin-8-yl).
  • A is an optionally substituted imidazo[1,2-a]pyridine-8-yl (e.g., unsubstituted imidazo[1,2-a]pyridine-8-yl).
  • A is an optionally substituted thiazolo[5,4-b]pyridin-7-yl (e.g., unsubstituted thiazolo[5,4-b]pyridine-7-yl).
  • A is an optionally substituted 1H-pyrrolo[3,2-b]pyridin-3-yl (e.g., unsubstituted 1H-pyrrolo[3,2-b]pyridin-3-yl).
  • A is an optionally substituted 1H-pyrrolo[2,3-b]pyridin-1-yl (e.g., unsubstituted 1H-pyrrolo[2,3-b]pyridin-1-yl).
  • A is an optionally substituted benzo[d]oxazol-2(3H)-on-6-yl (e.g., 3-methylbenzo[d]oxazol-2(3H)-on-6-yl).
  • A is an optionally substituted benzo[c]isothiazol-7-yl (e.g., unsubstituted benzo[c]isothiazol-7-yl).
  • A is aryl or heteroaryl, wherein the aryl or heteroaryl is optionally substituted, and the aryl or heteroaryl is fused to an optionally substituted carbocyclyl or an optionally substituted heterocyclyl;
  • R 1b is C 1-8 alkyl optionally substituted with one to four R 5 groups; C 1-8 alkenyl optionally substituted with one to four R 5 groups; cycloalkyl; heterocycle; aryl; heteroaryl; cycloalkylalkyl; cycloalkylalkenyl; heterocyclylalkyl; heterocyclylalkenyl; aralkyl; aralkenyl; heteroaralkyl; heteroaralkenyl; or —OH, with the proviso that when R 1a is OH, R 1b is not OH; wherein each cycloalkyl, heterocycle, aryl, heteroaryl, cycloalkylalkyl, cycloalkylalkenyl, heterocyclylalkyl, heterocyclylalkenyl, aralkyl, aralkenyl, heteroaralkyl, or heteroaralkenyl is optionally substituted;
  • each R 2 is independently selected from halo, alkyl, CN, OH, and alkoxy, wherein said alkyl or alkoxy is optionally substituted with one to four R 5 groups; or
  • R 2 groups are taken together with the ring atoms they are attached to form a 5- or 6-membered carbocyclic, aryl, heterocyclic or heteroaryl ring;
  • each R 4 is independently selected from halo, alkyl, alkoxy, haloalkyl, haloalkoxy and hydroxyl;
  • each R 5 is independently selected from halo, OH, C 1-6 alkoxy, CN, NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl) 2 ;
  • n 0, 1, 2 or 3;
  • n 0, 1 or 2.
  • X is —NH—S(O) 2 —, —NH—S(O) 2 —CH 2 —, or —CH 2 —S(O) 2 —NH—.
  • X is —NH—S(O) 2 —.
  • A is an optionally substituted quinolin-8-yl and X is —NH—S(O) 2 — and the compound has the structure set forth in formula (II) or a pharmaceutically acceptable salt thereof:
  • R 1b , R 2 , R 4 , m and n are as defined for Formula (I).
  • A is an optionally substituted quinolin-8-yl and X is —NH—S(O) 2 — and the compound has the structure set forth in formula (IIa) or a pharmaceutically acceptable salt thereof:
  • R 1b , R 2 , and n are as defined for Formula (Ia).
  • X is —NH—S(O) 2 —, —NH—S(O) 2 —CH 2 —, or —CH 2 —S(O) 2 —NH—.
  • X is —NH—S(O) 2 —.
  • A is an optionally substituted quinoxalin-5-yl and X is —NH—S(O) 2 — and the compound has the structure set forth in formula (III) or a pharmaceutically acceptable salt thereof:
  • R 1b , R 2 , R 4 , m and n are as defined for Formula (I).
  • A is an optionally substituted quinoxalin-5-yl and X is —NH—S(O) 2 — and the compound has the structure set forth in formula (IIIa) or a pharmaceutically acceptable salt thereof:
  • R 1b , R 2 , and n are as defined for Formula (Ia).
  • X is —NH—S(O) 2 —, —NH—S(O) 2 —CH 2 —, or —CH 2 —S(O) 2 —NH—.
  • X is —NH—S(O) 2 —.
  • A is an optionally substituted benzo[d]thiazol-4-yl and X is —NH—S(O) 2 — and the compound has the structure set forth in formula (III) or a pharmaceutically acceptable salt thereof:
  • R 1b , R 2 , R 4 , m and n are as defined for Formula (I).
  • A is an optionally substituted benzo[d]thiazol-4-yl and X is —NH—S(O) 2 — and the compound has the structure set forth in formula (IIIa) or a pharmaceutically acceptable salt thereof:
  • R 1b , R 2 , and n are as defined for Formula (Ia).
  • A is an optionally substituted monocyclic aryl (e.g., optionally substituted phenyl).
  • A is 4-chlorophenyl.
  • A is 3-cyanophenyl.
  • A is 2-chlorophenyl.
  • A is 4-cyanophenyl.
  • A is 2-trifluoromethylphenyl.
  • A is 4-trifluoromethylphenyl.
  • A is 3-trifluoromethylphenyl.
  • A is 3-chlorophenyl.
  • A is 4-trifluoromethoxyphenyl.
  • A is 2,3-dichlorophenyl.
  • A is 2,4-difluorophenyl.
  • A is 3-trifluoromethoxyphenyl.
  • A is phenyl substituted with two substituents on adjacent carbons which form an optionally substituted heterocyclyl or carbocyclyl ring (e.g., resulting in A comprising a bicycle).
  • R 1a is hydroxyl. In some embodiments of formula (I), R 1a is —C(O)H. In some embodiments of formula (I), R 1a is —CH 2 OH. In some embodiments of formula (I), R 1a is —CO 2 —C 1-6 alkyl (e.g., —CO 2 Et). In some embodiments of formula (I), R 1a is —CO 2 H. In some embodiments of formula (I), R 1a is —N(R 10a ) 2 . In some embodiments of formula (I), R 1a is —OP( ⁇ O)(OH) 2 . In some embodiments of formula (I), R 1a is —OCO 2 —CH 2 —OP( ⁇ O)(OH) 2 .
  • R 1b is cycloalkyl, heterocycle, aryl, heteroaryl, cycloalkylalkyl, cycloalkylalkenyl, heterocyclylalkyl, heterocyclylalkenyl, aralkyl, aralkenyl, heteroaralkyl, or heteroaralkenyl, wherein each cycloalkyl, heterocycle, aryl, heteroaryl, cycloalkylalkyl, cycloalkylalkenyl, heterocyclylalkyl, heterocyclylalkenyl, aralkyl, aralkenyl, heteroaralkyl, or heteroaralkenyl is optionally substituted with halo, C 1-6 alkyl, —OH, C 1-6 alkoxy, —CN, —NH 2 , —SO 2
  • R 1b is aryl; heteroaryl; aralkyl; or heteroaralkyl wherein each aryl; heteroaryl; aralkyl; or heteroaralkyl is optionally substituted with halo, C 1-6 alkyl, —OH, C 1-6 alkoxy, —CN, —NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), —N(C 1-6 alkyl) 2 , aryl, haloalkyl, or haloalkoxy.
  • R 1b is optionally substituted aralkyl (e.g., benzyl, 2,3-difluorobenzyl, 2-fluorobenzyl, 3-fluorobenzyl, 2-methylbenzyl, 3-methylbenzyl, 2-trifluoromethylbenzyl, 3-trifluoromethylbenzyl, 2-methoxybenzyl, 3-methoxybenzyl).
  • aralkyl e.g., benzyl, 2,3-difluorobenzyl, 2-fluorobenzyl, 3-fluorobenzyl, 2-methylbenzyl, 3-methylbenzyl, 2-trifluoromethylbenzyl, 3-trifluoromethylbenzyl, 2-methoxybenzyl, 3-methoxybenzyl.
  • R 1b is optionally substituted aryl (e.g., unsubstituted phenyl, 2-(2-chlorophenyl)phenyl, 4-cyanophenyl, 3-cyanophenyl, 2-cyanophenyl, 2-hydroxyphenyl, 2-(methylsulfonyl)phenyl, 3-(methylsulfonyl)phenyl, 2-chloro-4-methylphenyl, 2-chloro-4-fluorophenyl, 2-chloro-3-fluorophenyl, 2-chloro-5-fluorophenyl, 2,3-difluorophenyl, 2,6-difluorophenyl, 2-methylphenyl, 2-fluorophenyl, 2-methoxyphenyl, 2-trifluoromethylphenyl, 2-difluorophenyl, 3-
  • R 1b is phenyl, optionally substituted with chloro, fluoro, bromo, methyl, ethyl, —CN, difluoromethyl, trifluoromethyl, —OCF 3 , —SO 2 —CH 3 , or —OCH 3 .
  • R 1b is optionally substituted heteroaralkyl (e.g., methyl-3-pyridazinyl, methyl-3-pyridyl, methyl-2-pyridyl, 3-methyl-methyl-2-pyridyl, 2-fluoro-methyl-3-pyridyl or 3-fluoro-methyl-2-pyridyl).
  • heteroaralkyl e.g., methyl-3-pyridazinyl, methyl-3-pyridyl, methyl-2-pyridyl, 3-methyl-methyl-2-pyridyl, 2-fluoro-methyl-3-pyridyl or 3-fluoro-methyl-2-pyridyl.
  • R 1b is optionally substituted heteroaryl (e.g., 3-fluoro-2-pyridyl, 3-methyl-2-pyridyl, 3-fluoro-4-pyridyl, 4-pyridyl, 4-isothiazolyl, 3-methyl-4-fluoro-2-pyridyl, 2-chloro-4-pyridyl, 4-fluoro-2-methyl-3-pyridyl, 4-fluoro-3-bromo-2-pyridyl, 2-methoxy-3-pyridyl, 6-methoxy-2-pyridyl, 6-fluoro-2-pyridyl, 6-methyl-2-pyridyl, 2-methyl-3-pyridyl, 6-chloro-2-pyridyl, 3-trifluoromethyl-2-pyridyl, 6-trifluoromethyl-2-pyridyl, 6-trifluoromethyl-2-pyridyl, 6-trifluoromethyl-2-pyridyl, 6-trifluoro
  • R 1b is optionally substituted C 1-8 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, t-butyl, isobutyl, n-butyl, t-pentyl, 2-hydroxyethyl, 1-hydroxyethyl, 3-hydroxypropyl, 2-hydroxy-2-methylpropyl, 3-hydroxy-3-methylbutyl, 3,3,3-trifluoropropyl, 2-methoxyethyl, 3,3-difluoropropyl, ethoxymethyl, N,N-dimethylmethyl, pyrrollomethyl or 2-hydroxypropyl).
  • C 1-8 alkyl e.g., methyl, ethyl, n-propyl, isopropyl, t-butyl, isobutyl, n-butyl, t-pentyl, 2-hydroxyethyl, 1-hydroxyethyl, 3-hydroxypropyl, 2-hydroxy
  • R 1b is optionally substituted cycloalkyl (e.g., cyclopropyl).
  • R 1b is optionally substituted cycloalkylalkyl (e.g., cyclopropylmethyl, 1-methyl-cyclopropylmethyl, cyclobutylmethyl, 2,2-difluorocyclopropylmethyl).
  • R 1b is optionally substituted C 2-8 alkenyl (e.g., 2-methyl-2-propenyl or 3,3-difluoro-2-propenyl).
  • R 1b is C 1-8 alkyl optionally substituted with one to four R 5 groups (e.g., halo, —OH, C 1-6 alkoxy, CN, NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl) 2 ).
  • R 5 groups e.g., halo, —OH, C 1-6 alkoxy, CN, NH 2 , —SO 2 —C 1-6 alkyl, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl) 2 ).
  • R 1b is C 1-8 alkyl optionally substituted with one to four R 5 groups (e.g., chloro, fluoro, —OH, —OCH 3 , —OCH 2 CH 3 , —CN, —NH 2 , —SO 2 —CH 3 , —SO 2 —CH 2 CH 3 , —NH(CH 3 ), —N(CH 3 ) 2 , and —N(CH 3 )(CH 2 CH 3 )).
  • R 5 groups e.g., chloro, fluoro, —OH, —OCH 3 , —OCH 2 CH 3 , —CN, —NH 2 , —SO 2 —CH 3 , —SO 2 —CH 2 CH 3 , —NH(CH 3 ), —N(CH 3 ) 2 , and —N(CH 3 )(CH 2 CH 3 )).
  • R 1b is —NH—R 5 .
  • R 5 is optionally substituted aryl (e.g., 2-methoxyphenyl).
  • R 5 is optionally substituted aralkyl (e.g., unsubstituted benzyl).
  • R 1b is —NH—C(O)—R 5 .
  • R 5 is optionally substituted heteroaryl (e.g., unsubstituted 2-pyridyl).
  • R 1b is:
  • the compound is selected from any one of the compounds set forth in Table 1, below:
  • PKR mutants having lower activities compared to the wild type, thus are useful for methods of the present invention.
  • Such mutations in PKR can affect enzyme activity (catalytic efficiency), regulatory properties (modulation by fructose bisphosphate (FBP)/ATP), and/or thermostability of the enzyme. Examples of such mutations are described in Valentini et al, JBC 2002.
  • Some examples of the mutants that are activated by the compounds described herein include G332S, G364D, T384M, G37E, R479H, R479K, R486W, R532W, R510Q, and R490W.
  • compounds described herein affect the activities of PKR mutants by activating FBP non-responsive PKR mutants, restoring thermostability to mutants with decreased stability, or restoring catalytic efficiency to impaired mutants.
  • the activating activity of the present compounds against PKR mutants may be tested following a method described in Example 8.
  • Compounds described herein are also useful as activators of wild type PKR.
  • a compound, composition or pharmaceutical composition described herein is added directly to whole blood or packed cells extracorporeally or be provided to the patient directly (e.g., by i.p., i.v., i.m., oral, inhalation (aerosolized delivery), transdermal, sublingual and other delivery routes).
  • compounds described herein increase the lifetime of the RBCs, thus counteract aging of stored blood, by impacting the rate of release of 2,3-DPG from the blood.
  • a decrease in the level of 2, 3-DPG concentration induces a leftward shift of the oxygen-hemoglobin dissociation curve and shifts the allosteric equilibribrium to the R, or oxygenated state, thus producing a therapeutic inhibition of the intracellular polymerization that underlies sickling by increasing oxygen affinity due to the 2,3-DPG depletion, thereby stabilizing the more soluble oxy-hemoglobin.
  • compounds and pharmaceutical compositions described herein are useful as antisickling agents.
  • a compound, composition or pharmaceutical composition described herein is added directly to whole blood or packed cells extracorporeally or be provided to the patient directly (e.g., by i.p., i.v., i.m., oral, inhalation (aerosolized delivery), transdermal, sublingual and other delivery routes).
  • a compound described herein may be an activator of a PKR, for example, a wild type (wt), mutated PKR (e.g., R510Q, or R532W).
  • a wild type (wt) e.g., wt
  • mutated PKR e.g., R510Q, or R532W.
  • Activities of exemplary compounds against wt PKR (in an enzymatic or cell based assay) and mutant PKRs are shown in Table 2 as measured by assays in Examples 2-5 below.
  • AA refers to an AC 50 less than 100 nM
  • BB refers to an AC 50 from 101 nM to 1.00 ⁇ M
  • CC refers to an AC 50 from than 1.01 ⁇ M to 10.00 ⁇ M
  • DD refers to an AC 50 greater than 10.01 ⁇ M
  • EE refers to an AC50 that is not available.
  • the compounds provided herein may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are expressly included within the scope. Unless otherwise indicated when a compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.
  • the compounds provided herewith may also contain linkages (e.g., carbon-carbon bonds) or substituents that can restrict bond rotation, e.g., restriction resulting from the presence of a ring or double bond. Accordingly, all cis/trans and E/Z isomers are expressly included.
  • the compounds provided herein may also comprise one or more isotopic substitutions.
  • H may be in any isotopic form, including 1 H, 2 H (D or deuterium), and 3 H (T or tritium);
  • C may be in any isotopic form, including 12 C, 13 C, and 14 C;
  • O may be in any isotopic form, including 16 O and 18 O; and the like.
  • the compounds provided herein may also be represented in multiple tautomeric forms, in such instances, expressly includes all tautomeric forms of the compounds described herein, even though only a single tautomeric form may be represented (e.g., alkylation of a ring system may result in alkylation at multiple sites; all such reaction products are expressly included). All such isomeric forms of such compounds are expressly included. All crystal forms of the compounds described herein are expressly included.
  • a salt for example, can be formed between an anion and a positively charged substituent (e.g., amino) on a compound described herein. Suitable anions include chloride, bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, and acetate. Likewise, a salt can also be formed between a cation and a negatively charged substituent (e.g., carboxylate) on a compound described herein.
  • a positively charged substituent e.g., amino
  • Suitable anions include chloride, bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, and acetate.
  • a salt can also be formed between a cation and a negatively charged substituent (e.g., carboxylate) on a compound described herein.
  • Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion.
  • Examples of prodrugs include esters and other pharmaceutically acceptable derivatives, which, upon administration to a subject, are capable of providing active compounds.
  • the compounds provided herein may be modified by appending appropriate functionalities to enhance selected biological properties, e.g., targeting to a particular tissue.
  • modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • the compounds described herein may be used as platforms or scaffolds that may be utilized in combinatorial chemistry techniques for preparation of derivatives and/or chemical libraries of compounds.
  • Such derivatives and libraries of compounds have biological activity and are useful for identifying and designing compounds possessing a particular activity.
  • Combinatorial techniques suitable for utilizing the compounds described herein are known in the art as exemplified by Obrecht, D. and Villalgrodo, J.
  • one embodiment relates to a method of using the compounds described herein for generating derivatives or chemical libraries comprising: 1) providing a body comprising a plurality of wells; 2) providing one or more compounds identified by methods described herein in each well; 3) providing an additional one or more chemicals in each well; 4) isolating the resulting one or more products from each well.
  • An alternate embodiment relates to a method of using the compounds described herein for generating derivatives or chemical libraries comprising: 1) providing one or more compounds described herein attached to a solid support; 2) treating the one or more compounds identified by methods described herein attached to a solid support with one or more additional chemicals; 3) isolating the resulting one or more products from the solid support.
  • tags or identifier or labeling moieties may be attached to and/or detached from the compounds described herein or their derivatives, to facilitate tracking, identification or isolation of the desired products or their intermediates.
  • moieties are known in the art.
  • the chemicals used in the aforementioned methods may include, for example, solvents, reagents, catalysts, protecting group and deprotecting group reagents and the like. Examples of such chemicals are those that appear in the various synthetic and protecting group chemistry texts and treatises referenced herein.
  • the compounds described herein can be evaluated for ability to modulate PKM2 (e.g., activate PKM2) by methods known in the art.
  • compounds described herein are evaluated for ability to modulate PKM2 (e.g., activate PKM2) in serine deficient conditions.
  • exemplary methods include contacting the compound with a cell-based assay which allows assessment of the ability to modulate (e.g., activate) PKM2.
  • the candidate compound can be contacted with a cell and measuring the consumption of oxygen or production of lactate.
  • a change in cellular phosphoenolpyruvate, a change in glycerol-phosphate, a change in ribose or deoxyribose, a change in lipid synthesis, or a change in glucose conversion to lipid or nucleic acids or amino acids or protein can also be used to evaluate a compound for its ability to modulate PKM2 (e.g., activate PKM2).
  • the evaluation could also include measuring a change in pyruvate or a determination of an alteration in mitochondrial membrane potential, e.g., as measured by fluorescent potentiometric dyes.
  • PKM1 and PKM2 for use in the screening/testing method may be produced by any method known in the art for expression of recombinant proteins.
  • nucleic acids that encode the desired polypeptide may be introduced into various cell types or cell-free systems for expression.
  • Eukaryotic e.g., COS, HEK293T, CHO, and NIH cell lines
  • prokaryotic e.g., E. coli
  • a PKM sequence is introduced into a plasmid or other vector, which is then used to transform living cells.
  • PKM cDNA contains the entire open reading frame, or biologically active fragment thereof, are inserted in the correct orientation into an expression plasmid and may be used for protein expression.
  • Prokaryotic and eukaryotic expression systems allow for the expression and recovery of fusion proteins in which the PKM protein is covalently linked to a tag molecule on either the amino terminal or carboxy terminal side, which facilitates identification and/or purification.
  • tags that can be used include hexahistidine, HA, FLAG, and c-myc epitope tags.
  • An enzymatic or chemical cleavage site can be engineered between the PKM protein and the tag molecule so that the tag can be removed following purification.
  • the activity of the PKM enzyme measured in the screening/testing assay may be measured by, e.g., monitoring the concentration of a substrate (e.g., ATP or NADH) present in the reaction mixture.
  • a substrate e.g., ATP or NADH
  • Pyruvate produced by the enzymatic activity of pyruvate kinase, is converted into lactate by lactate dehydrogenase, which requires the consumption of NADH (NADH ⁇ NAD+).
  • the activity of PKM2 can be indirectly measured by monitoring the consumption of NADH through, e.g., fluorescence assays.
  • the activity of the PKM2 enzyme can be directly monitored by measuring the production of ATP, as ATP is produced when phosphoenolpyruvate is converted to pyruvate.
  • Methods for monitoring the amount of substrate in a reaction mixture include, e.g., absorbance, fluorescence, Raman scattering, phosphorescence, luminescence, luciferas
  • the screening procedure requires the presence of specific components in the reaction mixture.
  • Components utilized in the assay include, e.g., a nucleoside diphosphate (e.g., ADP), phosphoenolpyruvate, NADH, lactate dehydrogenase, FBP, a reducing agent (e.g., dithiothreitol), a detergent (e.g., Brij 35), glycerol, and a solvent (e.g., DMSO).
  • a nucleoside diphosphate e.g., ADP
  • phosphoenolpyruvate phosphoenolpyruvate
  • NADH e.g., lactate dehydrogenase
  • FBP e.g., FBP
  • a reducing agent e.g., dithiothreitol
  • a detergent e.g., Brij 35
  • glycerol e.g., glycerol
  • solvent
  • Compounds useful as PKM2 activators are those that demonstrate specificity and activation of PKM2 enzyme in the absence of FBP to a level greater than that of 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99, or 100% in the presence of FBP. Furthermore, compounds can be evaluated in the presence or absence of a phosphotyrosine peptide. Phosphotyrosine peptide binding to PKM2 leads to a dissociation of FBP from PKM2 and conformational changes of PKM2 from an active, tetrameric form to an inactive form.
  • a method for treating or preventing a disease, condition or disorder as described herein comprising administering a compound, a pharmaceutically acceptable salt of a compound or pharmaceutical composition comprising a compound described herein (e.g., a compound of formula (I), (Ia), (Ib), (II), (IIa), (III), (IIIa), (IV), (IVa), or in Table 1).
  • a compound described herein e.g., a compound of formula (I), (Ia), (Ib), (II), (IIa), (III), (IIIa), (IV), (IVa), or in Table 1).
  • the compounds and compositions described herein can be administered to cells in culture, e.g., in vitro or ex vivo, or to a subject, e.g., in vivo, to treat, prevent, and/or diagnose a variety of disorders, including those described herein below.
  • the term “treat” or “treatment” is defined as the application or administration of a compound, alone or in combination with, a second therapeutic agent to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a disorder (e.g., a disorder as described herein), a symptom of a disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder, or one or more symptoms of the disorder.
  • a disorder e.g., a disorder as described herein
  • an amount of a compound effective to treat a disorder refers to an amount of the compound which is effective, upon single or multiple dose administration to a subject, in treating a cell, or in curing, alleviating, relieving or improving a subject with a disorder beyond that expected in the absence of such treatment.
  • the term “prevent” is defined as the application or administration of a compound, alone or in combination with, a second therapeutic agent to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a predisposition toward a disorder, with the purpose to prevent the occurrence of at least one symptom of the disorder or to delay onset of at least one symptom of the disorder).
  • an amount of a compound effective to prevent a disorder refers to an amount effective, upon single- or multiple-dose administration to the subject, in preventing or delaying the occurrence of the onset or recurrence of a disorder or a symptom of the disorder.
  • the term “subject” is intended to include human and non-human animals.
  • exemplary human subjects include a human patient having a disorder, e.g., a disorder described herein or a normal subject.
  • non-human animals includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals, e.g., sheep, dog, cat, cow, pig, etc.
  • a compound or composition described herein can be used to treat a blood related condition.
  • a method for increasing lifetime of the red blood cells (RBCs) in need thereof comprising contacting blood with an effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a composition comprising a compound disclosed herein or a salt, solvate or hydrate thereof and a carrier; or (3) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for regulating 2,3-diphosphoglycerate levels in blood in need thereof comprising contacting blood with an effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a composition comprising a compound disclosed herein or a salt, solvate or hydrate thereof and a carrier; or (3) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating hereditary non-spherocytic haemolytic anemia comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating sickle cell anemia comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • hemolytic anemia e.g., chronic hemolytic anemia caused by phosphoglycerate kinase deficiency, Blood Cells Mol Dis, 2011; 46(3):206
  • administering comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating diseases or conditions that are associated with increased 2,3-diphosphoglycerate levels comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • thalassemia e.g., beta-thalassemia
  • hereditary spherocytosis hereditary elliptocytosis
  • abetalipoproteinemia or Bassen-Kornzweig syndrome
  • paroxysmal nocturnal hemoglobinuria acquired hemolytic anemia (e.g., congenital anemias (e.g., enzymopathies))
  • anemia of chronic diseases comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • a method for treating diseases or conditions that are associated with increased 2,3-diphosphoglycerate levels comprising administering to a subject in need thereof a therapeutically effective amount of (1) a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof; (2) a pharmaceutical composition comprising a compound disclosed herein or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • PKR mutants having lower activities compared to the wild type, thus are useful for methods of the present invention.
  • Such mutations in PKR can affect enzyme activity (catalytic efficiency), regulatory properties (modulation by fructose bisphosphate (FBP)/ATP), and/or thermostability of the enzyme. Examples of such mutations are described in Valentini et al, JBC 2002.
  • Some examples of the mutants that are activated by the compounds described herein include G332S, G364D, T384M, G37E, R479H, R479K, R486W, R532W, R510Q, and R490W.
  • compounds described herein affect the activities of PKR mutants by activating FBP non-responsive PKR mutants, restoring thermostability to mutants with decreased stability, or restoring catalytic efficiency to impaired mutants.
  • the activating activity of the present compounds against PKR mutants may be tested following a method described in Examples 2-5.
  • Compounds described herein are also activators of wild type PKR.
  • a compound, composition or pharmaceutical composition described herein is added directly to whole blood or packed cells extracorporeally or be provided to the subject (e.g., the patient) directly (e.g., by i.p., i.v., i.m., oral, inhalation (aerosolized delivery), transdermal, sublingual and other delivery routes).
  • compounds described herein increase the lifetime of the RBCs, thus counteract aging of stored blood, by impacting the rate of release of 2,3-DPG from the blood.
  • a decrease in the level of 2,3-DPG concentration induces a leftward shift of the oxygen-hemoglobin dissociation curve and shifts the allosteric equilibribrium to the R, or oxygenated state, thus producing a therapeutic inhibition of the intracellular polymerization that underlies sickling by increasing oxygen affinity due to the 2,3-DPG depletion, thereby stabilizing the more soluble oxy-hemoglobin.
  • compounds and pharmaceutical compositions described herein are useful as antisickling agents.
  • a compound or composition described herein can be used to treat a neoplastic disorder.
  • a “neoplastic disorder” is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth.
  • Exemplary neoplastic disorders include: carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
  • the disclosed methods are useful in the prevention and treatment of cancer, including for example, solid tumors, soft tissue tumors, and metastases thereof.
  • the disclosed methods are also useful in treating non-solid cancers.
  • Exemplary solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary.
  • Exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine.
  • a compound described herein is administered together with one or more additional cancer treatments.
  • exemplary cancer treatments include, for example: chemotherapy, targeted therapies such as antibody therapies, immunotherapy, and hormonal therapy. Examples of each of these treatments are provided below.
  • a compound described herein is administered with one or morechemotherapies.
  • Chemotherapy is the treatment of cancer with drugs that can destroy cancer cells. “Chemotherapy” usually refers to cytotoxic drugs which affect rapidly dividing cells in general, in contrast with targeted therapy. Chemotherapy drugs interfere with cell division in various possible ways, e.g., with the duplication of DNA or the separation of newly formed chromosomes. Most forms of chemotherapy target all rapidly dividing cells and are not specific for cancer cells, although some degree of specificity may come from the inability of many cancer cells to repair DNA damage, while normal cells generally can.
  • chemotherapeutic agents used in cancer therapy include, for example, antimetabolites (e.g., folic acid, purine, and pyrimidine derivatives) and alkylating agents (e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, toposimerase inhibitors and others).
  • antimetabolites e.g., folic acid, purine, and pyrimidine derivatives
  • alkylating agents e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, toposimerase inhibitors and others.
  • agents include Aclarubicin, Actinomycin, Alitretinon, Altretamine, Aminopterin, Aminolevulinic acid, Amrubicin, Amsacrine, Anagrelide, Arsenic trioxide, Asparaginase, Atrasentan, Belotecan, Bexarotene, endamustine, Bleomycin, Bortezomib, Busulfan, Camptothecin, Capecitabine, Carboplatin, Carboquone, Carmofur, Carmustine, Celecoxib, Chlorambucil, Chlormethine, Cisplatin, Cladribine, Clofarabine, Crisantaspase, Cyclophosphamide, Cytarabine, dacarbazine, Dactinomycin, Daunorubicin, Decitabine, Demecolcine, Docetaxel, Doxorubicin, Efaproxiral, Elesclomol, Elsamitrucin, Enocita
  • the chemotherapy agents can be used in combination with a compound described herein.
  • a compound described herein is administered with one or more targeted therapies.
  • Targeted therapy constitutes the use of agents specific for the deregulated proteins of cancer cells.
  • Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
  • Prominent examples are the tyrosine kinase inhibitors such as Axitinib, Bosutinib, Cediranib, dasatinib, erlotinib, imatinib, gefitinib, lapatinib, Lestaurtinib, Nilotinib, Semaxanib, Sorafenib, Sunitinib, and Vandetanib, and also cyclin-dependent kinase inhibitors such as Alvocidib and Seliciclib.
  • Monoclonal antibody therapy is another strategy in which the therapeutic agent is an antibody which specifically binds to a protein on the surface of the cancer cells.
  • Examples include the anti-HER2/neu antibody trastuzumab (HERCEPTIN®) typically used in breast cancer, and the anti-CD20 antibody rituximab and Tositumomab typically used in a variety of B-cell malignancies.
  • Other exemplary antibodies include Cetuximab, Panitumumab, Trastuzumab, Alemtuzumab, Bevacizumab, Edrecolomab, and Gemtuzumab.
  • Exemplary fusion proteins include Aflibercept and Denileukin diftitox.
  • the targeted therapy can be used in combination with a compound described herein.
  • Targeted therapy can also involve small peptides as “homing devices” which can bind to cell surface receptors or affected extracellular matrix surrounding the tumor. Radionuclides which are attached to these peptides (e.g., RGDs) eventually kill the cancer cell if the nuclide decays in the vicinity of the cell.
  • RGDs Radionuclides which are attached to these peptides
  • An example of such therapy includes BEXXAR®.
  • a compound described herein is administered with one or more immunotherapies.
  • Cancer immunotherapy refers to a diverse set of therapeutic strategies designed to induce the patient's own immune system to fight the tumor.
  • Contemporary methods for generating an immune response against tumors include intravesicular BCG immunotherapy for superficial bladder cancer, and use of interferons and other cytokines to induce an immune response in renal cell carcinoma and melanoma patients.
  • Allogeneic hematopoietic stem cell transplantation can be considered a form of immunotherapy, since the donor's immune cells will often attack the tumor in a graft-versus-tumor effect.
  • the immunotherapy agents can be used in combination with a compound described herein.
  • a compound described herein is administered with one or more hormonal therapies.
  • the growth of some cancers can be inhibited by providing or blocking certain hormones.
  • hormone-sensitive tumors include certain types of breast and prostate cancers. Removing or blocking estrogen or testosterone is often an important additional treatment.
  • administration of hormone agonists, such as progestogens may be therapeutically beneficial.
  • the hormonal therapy agents can be used in combination with a compound described herein.
  • a compound or composition described herein can be used to treat or prevent obesity, e.g., in a human subject, e.g., a child or adult subject.
  • “Obesity” refers to a condition in which a subject has a body mass index of greater than or equal to 30.
  • Many compounds described herein can be used to treat or prevent an over-weight condition.
  • “Over-weight” refers to a condition in which a subject has a body mass index of greater or equal to 25.0.
  • the body mass index (BMI) and other definitions are according to the “NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults” (1998).
  • Treatment with the compound may be in an amount effective to alter the weight of the subject, e.g., by at least 2, 5, 7, 10, 12, 15, 20, 25, 30, 25, 40, 45, 50, or 55%.
  • Treatment with a compound may be in an amount effective to reduce the body mass index of the subject, e.g., to less than 30, 28, 27, 25, 22, 20, or 18.
  • the compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
  • a compound or composition described herein can be administered to treat obesity associated with Prader-Willi Syndrome (PWS).
  • PWS is a genetic disorder associated with obesity (e.g., morbid obesity).
  • a compound or composition described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
  • reduce cholesterol e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol
  • reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
  • compositions delineated herein include the compounds delineated herein (e.g., a compound described herein), as well as additional therapeutic agents if present, in amounts effective for achieving a modulation of disease or disease symptoms, including those described herein.
  • pharmaceutically acceptable carrier or adjuvant refers to a carrier or adjuvant that may be administered to a patient, together with a compound provided herewith, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions provided herewith include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d- ⁇ -tocopherol polyethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-
  • Cyclodextrins such as ⁇ -, ⁇ -, and ⁇ -cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl- ⁇ -cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein.
  • compositions provided herewith may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection.
  • the pharmaceutical compositions provided herewith may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
  • surfactants such as Tweens or Spans and/or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions provided herewith may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions and/or emulsions are administered orally, the active ingredient may be suspended or dissolved in an oily phase is combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions provided herewith may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound provided herewith with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions provided herewith is useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds provided herewith include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier with suitable emulsifying agents.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions provided herewith may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches are also included.
  • compositions provided herewith may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • compositions provided herewith comprise a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds provided herewith. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds provided herewith in a single composition.
  • the compounds described herein can, for example, be administered by injection, intravenously, intraarterially, subdermally, intraperitoneally, intramuscularly, or subcutaneously; or orally, buccally, nasally, transmucosally, topically, in an ophthalmic preparation, or by inhalation, with a dosage ranging from about 0.5 to about 100 mg/kg of body weight, alternatively dosages between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug.
  • the methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect.
  • the pharmaceutical compositions provided herewith will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion.
  • Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w).
  • such preparations contain from about 20% to about 80% active compound.
  • a maintenance dose of a compound, composition or combination provided herewith may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • the compounds described herein can modulate PKM2. Accordingly, a patient and/or subject can be selected for treatment using a compound described herein by first evaluating the patient and/or subject to determine whether the subject is in need of modulation of PKM2, and if the subject is determined to be in need of modulation of PKM2, then administering to the subject a compound described herein.
  • a subject can be evaluated as being in need of modulation of PKM2 using methods known in the art, e.g., by measuring the presence and/or activity of PKM2 in the patient.
  • the activity and/or level of PKM2 is evaluated in the cancer.
  • a patient receiving a compound described herein can be monitored, for example, for improvement in the condition and/or adverse effects. Improvement of a patient's condition can be evaluated, for example, by monitoring the growth, absence of growth, or regression of the cancer (e.g., a tumor). In some embodiments, the patient is evaluated using a radiological assay or evaluation of hemolytic parameters.
  • the compounds described herein can activate mutant PKRs. Accordingly, a patient and/or subject can be selected for treatment using a compound described herein by first evaluating the patient and/or subject to determine whether the subject carries a mutation in PKR (for examples, one of the mutations as described herein), and if the subject is determined to be carrying a mutation in PKR thus is in need of activation of the activity of the mutant PKR, then optionally administering to the subject a compound described herein. A subject can be evaluated as carrying a mutation in PKR using methods known in the art.
  • the reagents were purchased from commercial sources (such as Alfa, Acros, Sigma Aldrich, TCI and Shanghai Chemical Reagent Company), and used without further purification.
  • Nuclear magnetic resonance (NMR) spectra were obtained on a Brucker AMX-400 NMR (Brucker, Switzerland). Chemical shifts were reported in parts per million (ppm, ⁇ ) downfield from tetramethylsilane.
  • Mass spectra were given with electrospray ionization (ESI) from a Waters LCT TOF Mass Spectrometer (Waters, USA). Microwave reactions were run on an Initiator 2.5 Microwave Synthesizer (Biotage, Sweden).
  • PKR mutant enzyme 80-400 ng/well; ADP: 0.22-1.65 mM; PEP: 0.1-0.5 mM; NADH:180 uM; LDH: 0.5 units (Sigma#59023); DTT: 1 mM; BSA: 0.03%.
  • a compound described herein was diluted with DMSO and tested at 1 ⁇ M concentration.
  • the enzyme was diluted in 1 ⁇ Buffer: (100 mM KCl, 50 mM Tris 7.5, 5 mM MgCl 2 , 1 mM DTT, 0.03% BSA). 2 ⁇ L of compound solution was first added into wells, and then 180 ⁇ L of enzyme solution was added. Assays were assembled except for ADP, and plates were stored for 60 minutes at RT. 20 ⁇ L ADP was added to start the assay and assay output was evaluated using OD340 at SpectraMax. The assay was run at room temperature.
  • PKR wt 100 ng/well
  • Tris pH 7.5 50 mM
  • KCl 100 mM
  • MgCl 2 5 mM
  • ADP 0.48 mM
  • PEP 0.15 mM
  • NADH 180 ⁇ M
  • LDH 0.5 units, Sigma 59023
  • DTT 1 mM
  • BSA 0.03%
  • a compound described herein was diluted with DMSO and tested at 1 ⁇ M concentration.
  • the enzyme was diluted in 1 ⁇ Buffer: (100 mM KCl, 50 mM Tris 7.5, 5 mM MgCl 2 , 1 mM DTT, 0.03% BSA). 2 ⁇ L of compound solution was first added into wells, and then 180 ⁇ L of enzyme solution was added. Assays were assembled except for ADP, and plates were stored for 60 minutes at RT. 20 ⁇ L ADP was added to start the assay and assay output was evaluated using OD340 at SpectraMax. The assay was run at room temperature.
  • PKR R510Q (40 ng/well), Tris pH 7.5 (50 mM), KCl (100 mM), MgCl 2 (5 mM), ADP (0.2 mM), PEP (0.11 mM), NADH (180 ⁇ M), LDH (0.5 units, Sigma 59023), DTT (1 mM) and BSA (0.03%).
  • a compound described herein was diluted with DMSO and tested at 1 ⁇ M concentration.
  • the enzyme was diluted in 1 ⁇ Buffer: (100 mM KCl, 50 mM Tris 7.5, 5 mM MgCl 2 , 1 mM DTT, 0.03% BSA). 2 ⁇ L of compound solution was first added into wells, and then 180 ⁇ L of enzyme solution was added. Assays were assembled except for ADP, and plates were stored for 60 minutes at RT. 20 ⁇ L ADP was added to start the assay and assay output was evaluated using OD340 at SpectraMax. The assay was run at room temperature.
  • PKR R532W 100 ng/well
  • Tris pH 7.5 50 mM
  • KCl 100 mM
  • MgCl 2 5 mM
  • ADP 0.36 mM
  • PEP 0.1 mM
  • NADH 180 ⁇ M
  • LDH 0.5 units, Sigma 59023
  • DTT 1 mM
  • BSA 0.03%
  • Step A 4-(quinoline-8-sulfonamido)benzoic acid (1A)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
US14/776,635 2013-03-15 2014-03-13 Therapeutic compounds and compositions Abandoned US20160046579A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CNPCT/CN2013/072688 2013-03-15
PCT/CN2013/072688 WO2014139144A1 (en) 2013-03-15 2013-03-15 Therapeutic compounds and compositions
PCT/CN2014/000260 WO2014139325A1 (en) 2013-03-15 2014-03-13 Therapeutic compounds and compositions

Publications (1)

Publication Number Publication Date
US20160046579A1 true US20160046579A1 (en) 2016-02-18

Family

ID=51535825

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/776,635 Abandoned US20160046579A1 (en) 2013-03-15 2014-03-13 Therapeutic compounds and compositions
US14/210,583 Active US9108921B2 (en) 2013-03-15 2014-03-14 Therapeutic compounds and compositions
US14/814,862 Active US9365545B2 (en) 2013-03-15 2015-07-31 Therapeutic compounds and compositions

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/210,583 Active US9108921B2 (en) 2013-03-15 2014-03-14 Therapeutic compounds and compositions
US14/814,862 Active US9365545B2 (en) 2013-03-15 2015-07-31 Therapeutic compounds and compositions

Country Status (22)

Country Link
US (3) US20160046579A1 (de)
EP (1) EP2970189B1 (de)
JP (2) JP6374413B2 (de)
CN (1) CN105121425A (de)
AR (1) AR095557A1 (de)
AU (1) AU2014231564B2 (de)
BR (1) BR112015023760A2 (de)
CA (1) CA2903067A1 (de)
CL (1) CL2015002767A1 (de)
CR (1) CR20150571A (de)
ES (1) ES2803548T3 (de)
HK (1) HK1216175A1 (de)
IL (1) IL241352A0 (de)
MX (1) MX2015012882A (de)
NI (1) NI201500140A (de)
PE (1) PE20151793A1 (de)
PH (1) PH12015502161A1 (de)
RU (1) RU2015143908A (de)
SG (1) SG11201507332SA (de)
TW (1) TW201444797A (de)
WO (2) WO2014139144A1 (de)
ZA (1) ZA201506487B (de)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014139144A1 (en) * 2013-03-15 2014-09-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
WO2016044463A2 (en) 2014-09-17 2016-03-24 Celgene Avilomics Research, Inc. Mk2 inhibitors and uses thereof
CN104817490B (zh) * 2015-05-13 2017-10-13 北京大学 氨基二硫代甲酸酯类化合物及其制备方法与应用
LT3386591T (lt) 2015-12-09 2020-10-12 Cadent Therapeutics, Inc. Heteroaromatiniai nmda receptoriaus moduliatoriai ir jų panaudojimas
EP4006038A1 (de) 2015-12-09 2022-06-01 Cadent Therapeutics, Inc. Thienopyrimidinon-nmda-rezeptor-modulatoren und verwendungen davon
GB201617758D0 (en) * 2016-10-20 2016-12-07 Almac Discovery Limited Pharmaceutical compounds
JP7332472B2 (ja) 2016-12-22 2023-08-23 ノバルティス アーゲー Nmda受容体モジュレーターおよびその使用
NZ748072A (en) * 2017-03-20 2020-06-26 Forma Therapeutics Inc Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators
CN107298685A (zh) * 2017-06-29 2017-10-27 上海合全药业股份有限公司 一种8‑(叔丁氧羰基)‑1‑氧杂‑8‑氮杂螺[4.5]癸烷‑2‑羧酸的合成方法
MX2020001833A (es) 2017-08-15 2020-07-22 Agios Pharmaceuticals Inc Moduladores de piruvato quinasas y uso de los mismos.
AU2018355743B2 (en) 2017-10-27 2022-04-14 Boehringer Ingelheim International Gmbh Pyridine carbonyl derivatives and therapeutic uses thereof as TRPC6 inhibitors
EP3604288B1 (de) 2018-08-02 2024-04-10 Development Center for Biotechnology Regioselektives einstufiges verfahren zur synthese von 2-hydroxychinoxalin
MX2021001367A (es) 2018-08-03 2021-06-23 Cadent Therapeutics Inc Moduladores del receptor nmda heteroaromático y usos de los mismos.
CN113226356A (zh) 2018-09-19 2021-08-06 福马治疗股份有限公司 活化丙酮酸激酶r
US12053458B2 (en) 2018-09-19 2024-08-06 Novo Nordisk Health Care Ag Treating sickle cell disease with a pyruvate kinase R activating compound
WO2020129877A1 (ja) * 2018-12-18 2020-06-25 株式会社デ・ウエスタン・セラピテクス研究所 イソキノリンスルホニルクロリド酸付加塩及びその製造方法
WO2021029450A1 (ko) * 2019-08-09 2021-02-18 한국화학연구원 신규한 피리미딘 설폰아마이드 유도체 및 이를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물
KR20230051247A (ko) 2020-08-13 2023-04-17 베링거 인겔하임 인터내셔날 게엠베하 정신분열병과 연관된 인지 손상의 치료
CN112773800B (zh) * 2021-01-25 2021-10-15 南通大学 哌啶-1-二硫代甲酸-3-甲基-1,4-二氧-1,4-二氢萘-2-甲基酯的用途
WO2022170200A1 (en) 2021-02-08 2022-08-11 Global Blood Therapeutics, Inc. 1-(2-sulfonyl-2,6-dihydropyrrolo[3,4-c]pyrazol-5(4h)-yl]-ethanone derivatives as pyruvate kinase (pkr) and pkm2 activators for the treatment of sickle cell disease
WO2023116774A1 (zh) 2021-12-21 2023-06-29 赛诺哈勃药业(成都)有限公司 含二氮杂亚基磺酰结构的化合物及其在医药上的用途
CN115073422A (zh) * 2022-07-28 2022-09-20 成都普瑞熙药业有限公司 一种制备4-(1-甲基-1h-吡唑-5-基)哌啶及其盐的方法
ES2975743A1 (es) * 2022-11-22 2024-07-12 Moehs Iberica Sl Metodo de preparacion de mitapivat, intermedios de sintesis de mitapivat y metodos de preparacion de los mismos

Family Cites Families (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE516129A (de) 1949-07-23
GB935538A (en) 1959-04-06 1963-08-28 Stop Motion Devices Corp Stop-motion head for use on knitting machines
US4474599A (en) 1982-07-14 1984-10-02 The Dow Chemical Company 1-(Pyridyl)-1H-1,2,3-triazole derivatives, and use as herbicidal agents
GB8325370D0 (en) 1983-09-22 1983-10-26 Fujisawa Pharmaceutical Co Benzoxazoline and benzothiazoline derivatives
JPS61129129A (ja) 1984-11-28 1986-06-17 Kureha Chem Ind Co Ltd 抗腫瘍剤
FI855180A (fi) 1985-01-18 1986-07-19 Nissan Chemical Ind Ltd Pyrazolsulfonamidderivat, foerfarande foer dess framstaellande och det innehaollande ograesgift.
EP0246749A3 (de) 1986-05-17 1988-08-31 AgrEvo UK Limited Herbizide Triazole
JPS6339875A (ja) 1986-08-05 1988-02-20 Nissin Food Prod Co Ltd ピリミジン誘導体
US4775762A (en) 1987-05-11 1988-10-04 The Dow Chemical Company Novel (1H-1,2,3-triazol-1-yl)pyridines
DE3813886A1 (de) 1988-04-20 1989-11-02 Schering Ag 1-triazinyl-1h-1,2,4-triazol-3- sulfonsaeureamide, verfahren zu ihrer herstellung und ihre verwendung als mittel mit herbizider, fungizider und pflanzenwachstumsregulierender wirkung
DE3813885A1 (de) 1988-04-20 1989-11-02 Schering Ag 1-chlorpyrimidinyl-1h-1,2,4-triazol-3-sulfonsaeureamide, verfahren zu ihrer herstellung und ihre verwendung als mittel mit herbizider, fungizider und pflanzenwachstumsregulierender wirkung
US5220028A (en) 1988-10-27 1993-06-15 Nissan Chemical Industries, Ltd. Halogeno-4-methylpyrazoles
MY106533A (en) 1990-02-20 1995-06-30 Sumitomo Chemical Co A 4-tert.-butyl imidazole derivative and its production and use.
CA2036147A1 (en) 1990-06-29 1991-12-30 Hiroki Tomioka A 1-pyridylimidazole derivative and its production and use
AU9152191A (en) 1990-12-31 1992-08-17 Monsanto Company Reducing pesticidal interactions in crops
AU644297B2 (en) 1991-06-28 1993-12-02 Sumitomo Chemical Company, Limited A 1-pyridylimidazole derivative and its production and use
JPH0625177A (ja) 1992-07-09 1994-02-01 Nissan Chem Ind Ltd ピラゾール誘導体及び除草剤
JP3719612B2 (ja) 1993-06-14 2005-11-24 塩野義製薬株式会社 ヘテロ環を含有する尿素誘導体
DE19541146A1 (de) 1995-10-25 1997-04-30 Schering Ag Imidazolderivate und deren Verwendung als Stickstoffmonoxid-Synthase-Inhibitoren
FR2744449B1 (fr) 1996-02-02 1998-04-24 Pf Medicament Nouvelles piperazines aromatiques derivees de cycloazanes substitues, ainsi que leur procede de preparation, les compositions pharmaceutiques et leur utilisation comme medicaments
BR9815453A (pt) 1997-03-11 2001-10-23 Du Pont Composto, composição herbicida e método para controle do crescimento de vegetação indesejada
US7863444B2 (en) 1997-03-19 2011-01-04 Abbott Laboratories 4-aminopyrrolopyrimidines as kinase inhibitors
DE19743435A1 (de) 1997-10-01 1999-04-08 Merck Patent Gmbh Benzamidinderivate
US6214879B1 (en) 1998-03-24 2001-04-10 Virginia Commonwealth University Allosteric inhibitors of pyruvate kinase
JP2002523451A (ja) 1998-09-01 2002-07-30 ブリストル−マイヤーズ スクイブ カンパニー カリウムチャネル抑制剤および方法
SK3852001A3 (en) 1998-09-18 2003-03-04 Basf Ag 4-Aminopyrrolopyrimidines as kinase inhibitors
US6492408B1 (en) 1999-07-21 2002-12-10 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
BR0013694A (pt) 1999-09-04 2002-05-21 Astrazeneca Ab Composto, processo para a preparação de um composto, composição farmacêutica, e, uso de um composto
JP2002193710A (ja) 2000-12-25 2002-07-10 Kumiai Chem Ind Co Ltd ピリミジン又はトリアジン誘導体及び農園芸用殺菌剤
EP1387679A4 (de) 2001-05-07 2004-08-11 Smithkline Beecham Corp Sulfonamide
MXPA04001290A (es) 2001-08-15 2004-05-27 Du Pont Arilamidas sustituidas en la posicion orto para controlar plagas de invertebrados.
JP2003081937A (ja) 2001-09-07 2003-03-19 Bayer Ag ベンゼンスルホンアミド誘導体
US7169788B2 (en) 2001-10-30 2007-01-30 Merck & Co., Inc. Tyrosine kinase inhibitors
EP1472248A1 (de) 2002-01-17 2004-11-03 Eli Lilly And Company Acetylcholinrezeptor-modulatoren
CN101450934B (zh) 2002-03-13 2012-10-10 詹森药业有限公司 用作组蛋白去乙酰酶抑制剂的磺酰基衍生物
WO2003093297A2 (en) 2002-05-03 2003-11-13 Exelixis, Inc. Protein kinase modulators and methods of use
GB0215775D0 (en) 2002-07-06 2002-08-14 Astex Technology Ltd Pharmaceutical compounds
CA2494962C (en) 2002-08-09 2011-06-14 Merck & Co., Inc. Tyrosine kinase inhibitors
MXPA05004434A (es) 2002-10-24 2005-07-26 Sterix Ltd Inhibidores de 11-beta-hidroxi esteroide deshidrogenasa tipo 1 y tipo 2.
CA2509711A1 (en) * 2002-12-13 2004-07-01 Smithkline Beecham Corporation Piperidine derivatives as ccr5 antagonists
ATE482747T1 (de) * 2003-04-11 2010-10-15 High Point Pharmaceuticals Llc Neue amide derivate und deren pharmazeutische verwendungen
CN1798733A (zh) 2003-06-10 2006-07-05 凯利普西斯公司 作为组蛋白脱乙酰基酶抑制剂的疾病治疗用羰基化合物
US6818631B1 (en) 2003-08-15 2004-11-16 Nippon Soda Co. Ltd. Fungicidal pyrimidine derivatives
US20080051414A1 (en) 2003-10-14 2008-02-28 Arizona Board Of Regents On Behalf Of The University Of Arizona Protein Kinase Inhibitors
WO2006033628A1 (en) 2004-09-24 2006-03-30 Astrazeneca Ab Benzimidazole derivatives, compositions containing them, preparation thereof and uses thereof
WO2006038594A1 (ja) * 2004-10-04 2006-04-13 Ono Pharmaceutical Co., Ltd. N型カルシウムチャネル阻害薬
WO2006043950A1 (en) 2004-10-20 2006-04-27 Smithkline Beecham Corporation Il-8 receptor antagonists
WO2006052546A2 (en) 2004-11-04 2006-05-18 Neurogen Corporation Pyrazolylmethyl heteroaryl derivatives
SE0402762D0 (sv) 2004-11-11 2004-11-11 Astrazeneca Ab Indazole sulphonamide derivatives
SI1879877T1 (sl) 2005-05-03 2013-02-28 Ranbaxy Laboratories Limited Protimikrobna sredstva
US20100105657A1 (en) 2005-07-05 2010-04-29 Astrazeneca Ab Compounds, Process for their Preparation, Intermediates, Pharmaceutical Compositions and their use in the Treatment of 5-HT6 Mediated Disorders such as Alzheimer's Disease, Cognitive Disorders, Cognitive Impairment Associated with Schizophrenia, Obesity and Parkinson's Disease
US20080214495A1 (en) 2005-07-08 2008-09-04 Astrazeneca Ab Heterocyclic Sulfonamide Derivatives as Inhibitors of Factor Xa
EP1910342A1 (de) 2005-07-29 2008-04-16 Kalypsys, Inc. Multicyclische sulfonamidverbindungen als histondeacetylase-inhibitoren zur krankheitsbehandlung
JP2009503117A (ja) 2005-08-04 2009-01-29 サートリス ファーマシューティカルズ, インコーポレイテッド サーチュインモジュレーターとしてのベンゾイミダゾール誘導体
JP2007238458A (ja) 2006-03-06 2007-09-20 D Western Therapeutics Institute Inc 新規なイソキノリン誘導体及びこれを含有する医薬
US20090054453A1 (en) 2006-03-17 2009-02-26 Lilian Alcaraz Novel Tetralins as 5-HT6 Modulators
EP2004619A1 (de) 2006-03-23 2008-12-24 Amgen Inc. Heterocyclische 1-phenylsulfonyl-diaza-amidverbindungen und ihre verwendung als modulatoren von hydroxsteroid-dehydrogenasen
JP5255559B2 (ja) 2006-03-31 2013-08-07 アボット・ラボラトリーズ インダゾール化合物
CA2659956C (en) 2006-08-04 2016-01-05 Lewis C. Cantley Inhibitors of pyruvate kinase and methods of treating disease
WO2008024284A2 (en) 2006-08-21 2008-02-28 Merck & Co., Inc. Sulfonylated piperazines as cannabinoid-1 receptor modulators
CA2661741C (en) 2006-09-01 2014-04-29 Otsuka Chemical Co., Ltd. N-pyridylpiperidine compound, method for producing the same, and pest control agent
ES2435430T3 (es) 2006-10-16 2013-12-19 Thesan Pharmaceuticals, Inc. Pirazolil tienopiridinas terapéuticas
HUP0600810A3 (en) 2006-10-27 2008-09-29 Richter Gedeon Nyrt New sulfonamide derivatives as bradykinin antagonists, process and intermediates for their preparation and pharmaceutical compositions containing them
EP1995241B1 (de) * 2007-03-23 2010-03-17 ICAgen, Inc. Ionenkanal-Hemmer
PA8790401A1 (es) 2007-07-18 2009-03-31 Janssen Pharmaceutica Nv Sulfonamidas como moduladores de trpm8
EP2053045A1 (de) 2007-10-26 2009-04-29 Syngenta Participations AG Neue Imidazolderivate
BRPI0818855A2 (pt) 2007-10-26 2015-04-14 Syngenta Participations Ag Derivados de imidazol
JP2011507910A (ja) * 2007-12-21 2011-03-10 ユニバーシティー オブ ロチェスター 真核生物の寿命を変更するための方法
KR20110053347A (ko) * 2008-08-05 2011-05-20 탈자진 인코포레이티드 탈라세미아를 치료하는 방법
US8623859B2 (en) * 2008-08-22 2014-01-07 Evotec Ag Bradykinin B1 antagonists
GB0815781D0 (en) * 2008-08-29 2008-10-08 Xention Ltd Novel potassium channel blockers
ES2620634T3 (es) 2008-10-09 2017-06-29 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Activadores de piruvato quinasa humana
CN102448951B (zh) 2009-04-06 2017-05-10 安吉奥斯医药品有限公司 丙酮酸激酶m2调节剂、治疗组合物和相关使用方法
BRPI1013538A2 (pt) 2009-04-22 2019-09-24 Janssen Pharmaceutica Nv azetidinil diamidas como inibidores de monoacilglicerol lipase
DK2427441T3 (en) 2009-05-04 2017-03-20 Agios Pharmaceuticals Inc PKM2 Activators for use in the treatment of cancer
WO2010130638A1 (en) * 2009-05-14 2010-11-18 Evotec Ag Sulfonamide compounds, pharmaceutical compositions and uses thereof
KR101850813B1 (ko) * 2009-06-29 2018-04-20 아지오스 파마슈티컬스 아이엔씨. 치료용 화합물 및 조성물
AU2011210567B2 (en) 2010-01-29 2015-04-16 Dana-Farber Cancer Institute, Inc. Small molecules for the modulation of MCL-1 and methods of modulatiing cell death, cell division, cell differentiation and methods of treating disorders
KR20130044382A (ko) 2010-03-01 2013-05-02 마이렉시스 인코포레이티드 화합물 및 그의 치료 용도
AU2011245441B2 (en) 2010-04-29 2014-12-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Activators of human pyruvate kinase
DE102010048800A1 (de) 2010-10-20 2012-05-10 Merck Patent Gmbh Chinoxalinderivate
FR2967674B1 (fr) 2010-11-23 2012-12-14 Pf Medicament Derives d'heteroarylsulfonamides, leur preparation et leur application en therapeutique humaine
ES2564952T3 (es) * 2010-12-17 2016-03-30 Agios Pharmaceuticals, Inc. Nuevos derivados de N-(4-(azetidina-1-carbonil)fenil)-(hetero-)arilsulfonamida como moduladores de piruvato quinasa M2 (PKM2)
JP6267112B2 (ja) * 2011-05-03 2018-01-24 アジオス ファーマシューティカルズ, インコーポレイテッド ピルビン酸キナーゼアクチベーターの使用方法
CN102805860A (zh) * 2011-05-30 2012-12-05 复旦大学 丙酮酸激酶2乙酰化抑制剂及其用途
WO2014139144A1 (en) * 2013-03-15 2014-09-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions

Also Published As

Publication number Publication date
SG11201507332SA (en) 2015-10-29
AU2014231564A1 (en) 2015-09-10
US9108921B2 (en) 2015-08-18
IL241352A0 (en) 2015-11-30
CA2903067A1 (en) 2014-09-18
EP2970189A4 (de) 2016-08-10
AR095557A1 (es) 2015-10-28
HK1216175A1 (zh) 2016-10-21
WO2014139325A1 (en) 2014-09-18
WO2014139144A1 (en) 2014-09-18
AU2014231564B2 (en) 2018-07-12
EP2970189B1 (de) 2020-05-06
TW201444797A (zh) 2014-12-01
US20140288081A1 (en) 2014-09-25
AU2014231564A2 (en) 2015-10-22
CR20150571A (es) 2016-01-06
PE20151793A1 (es) 2015-12-10
CN105121425A (zh) 2015-12-02
ES2803548T3 (es) 2021-01-27
JP2016512203A (ja) 2016-04-25
NI201500140A (es) 2015-11-24
JP6374413B2 (ja) 2018-08-15
BR112015023760A2 (pt) 2017-07-18
JP2017105832A (ja) 2017-06-15
RU2015143908A (ru) 2017-04-21
US20150336931A1 (en) 2015-11-26
MX2015012882A (es) 2015-12-03
ZA201506487B (en) 2016-11-30
CL2015002767A1 (es) 2016-12-30
EP2970189A1 (de) 2016-01-20
PH12015502161A1 (en) 2016-01-25
US9365545B2 (en) 2016-06-14

Similar Documents

Publication Publication Date Title
US9365545B2 (en) Therapeutic compounds and compositions
US9458132B2 (en) Therapeutic compounds and compositions and their use as PKM2 modulators
US9221792B2 (en) N-(4-(azetidine-1-carbonyl) phenyl)-(hetero-) arylsulfonamide derivatives as pyruvate kinase M2 (PMK2) modulators
US20170001990A1 (en) Piperidine-dione derivatives
EP3793989B1 (de) Thiophenderivate
JP6527520B2 (ja) ピルビン酸脱水素酵素キナーゼの阻害剤としてのn1−(3,3,3−トリフルオロ−2−ヒドロキソ−2−メチルプロピオニル)−ピペリジン誘導体
CA2920537C (en) Piperidine urea derivatives as inhibitors of tankyrase
KR20150131308A (ko) 치료 화합물 및 조성물
US20220096454A1 (en) Carboxylic acid derivatives

Legal Events

Date Code Title Description
AS Assignment

Owner name: AGIOS PHARMACEUTICALS, INC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CIANCHETTA, GIOVANNI;POPOVICI-MULLER, JANETA;ZAHLER, ROBERT;SIGNING DATES FROM 20151209 TO 20151212;REEL/FRAME:037922/0107

Owner name: AGIOS PHARMACEUTICALS, INC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VIVA BIOTECH (SHANGHAI) LTD.;REEL/FRAME:038038/0586

Effective date: 20151217

Owner name: VIVA BIOTECH (SHANGHAI) LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CAO, SHELDON;WANG, XIAOLEI;YE, ZHIXIONG;SIGNING DATES FROM 20151214 TO 20151216;REEL/FRAME:038038/0579

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION