WO2016004418A1 - Thérapie par inhibiteur de glutaminase - Google Patents

Thérapie par inhibiteur de glutaminase Download PDF

Info

Publication number
WO2016004418A1
WO2016004418A1 PCT/US2015/039153 US2015039153W WO2016004418A1 WO 2016004418 A1 WO2016004418 A1 WO 2016004418A1 US 2015039153 W US2015039153 W US 2015039153W WO 2016004418 A1 WO2016004418 A1 WO 2016004418A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
nrf2
subject
inhibitor
gls
Prior art date
Application number
PCT/US2015/039153
Other languages
English (en)
Inventor
Timothy Heffernan
Carlo Toniatti
Jeffrey Kovacs
Virginia GIULIANI
Nakia SPENCE
Maria Emilia Di Francesco
Christopher A. BRISTOW
Original Assignee
Board Of Regents, University Of Texas System
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
Priority claimed from US14/791,206 external-priority patent/US20160002248A1/en
Priority claimed from US14/791,186 external-priority patent/US9809588B2/en
Application filed by Board Of Regents, University Of Texas System filed Critical Board Of Regents, University Of Texas System
Priority to ES15814655T priority Critical patent/ES2921989T3/es
Priority to EP15814655.5A priority patent/EP3164195B1/fr
Priority to DK15814655.5T priority patent/DK3164195T3/da
Priority claimed from US14/791,284 external-priority patent/US20160002204A1/en
Publication of WO2016004418A1 publication Critical patent/WO2016004418A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants

Definitions

  • Glutamine the most abundant amino acid in circulation, plays an essential role in providing cancer cells with biosynthetic intermediates required to support proliferation and survival. Specifically, glutaminolysis, or the enzymatic conversion of glutamine to glutamate, provides proliferating cancer cells with a source of nitrogen for amino acid and nucleotide synthesis, and a carbon skeleton to fuel ATP and NADPH synthesis through the TCA cycle.
  • glutamine metabolism plays a critical role in maintaining cellular redox homeostasis as glutamate can be converted into glutathione, the major intracellular antioxidant.
  • Cancer cells require a constant source of biomass and macro-molecules to support cell division and reducing agents to maintain redox homeostasis. While many of these building blocks are provided through aerobic glycolysis, many cancer cells have evolved a dependence on glutamine metabolism for growth and survival.
  • Glutamine metabolism i.e., glutaminolysis is regulated by mitochondrial glutaminase (GLS), the rate limiting enzyme that catalyzes the conversion of glutamine to glutamate and ammonia.
  • GLS mitochondrial glutaminase
  • Mammalian cells contain two genes that encode glutaminase: the kidney-type (GLS-1) and liver-type (GLS-2) enzymes. Each has been detected in multiple tissue types, with GLS-1 being widely distributed throughout the body.
  • GLS-1 is a phosphate-activated enzyme that exists in humans as two major splice variants, a long form (referred to as KGA) and a short form (GAC), which differ only in their C- terminal sequences.
  • GLS-1 Both forms of GLS-1 are thought to bind to the inner membrane of the mitochondrion in mammalian cells, although at least one report suggests that glutaminase may exist in the intramembrane space, dissociated from the membrane. GLS is frequently overexpressed in human tumors and has been shown to be positively regulated by oncogenes such as Myc. Consistent with the observed dependence of cancer cell lines on glutamine metabolism, pharmacological inhibition of GLS offers the potential to target Gin addicted tumors. Such targeted treatment, however, is hampered by the lack of clinical biomarkers to identify sensitive patient populations.
  • the invention is based, in part, on the discovery of biomarkers and
  • the invention comprises evaluating whether to administer a compound that inhibits glutathione production to a subject by determining the presence of certain biomarkers or mechanisms and administering such a compound to the patient.
  • the invention also comprises methods of treating patients identified by the presence of the biomarkers or mechanisms.
  • the invention comprises a method of treatment of a subject having a disorder in need of treatment comprising administering a compound that inhibits glutathione production to a subject.
  • the method comprises optionally obtaining a biological sample from a subject, determining that the
  • NRF2/KEAP1 pathway in said subject is deregulated, or that NRF2 signaling in said subject is hyperactive, or determining the presence of a loss-of-function mutation in KEAP1 or a gain-of-function mutation in NRF2 of said subject, or an increase in the intracellular concentration of glutathione in said subject and determining that the compound that inhibits glutathione production should be administered to the subject.
  • the invention comprises a method of treating a subject in need of treatment.
  • the method comprises determining that the NRF2/KEAP1 pathway in said subject is deregulated, that NRF2 signaling in said subject is hyperactive, that the nucleic acid of said subject comprises a loss-of-function mutation in KEAP1 or a gain-of-function mutation in NRF2, or determining an increase in the intracellular concentration of glutathione in said subject and administering a glutaminase inhibitor to the subject.
  • the subject in need of treatment may be afflicted with a disorder or a condition, for example, cancer, including, but not limited to, bladder cancer, breast cancer, bone marrow cancer, cancer of the central nervous system, cervical cancer, colon cancer, endometrial cancer, cancer of the gastric system, head and neck cancer, kidney cancer, liver cancer, lung cancer, muscle cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, or thyroid cancer.
  • a disorder or a condition for example, cancer, including, but not limited to, bladder cancer, breast cancer, bone marrow cancer, cancer of the central nervous system, cervical cancer, colon cancer, endometrial cancer, cancer of the gastric system, head and neck cancer, kidney cancer, liver cancer, lung cancer, muscle cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, or thyroid cancer.
  • the glutaminase inhibitor may, for example, be a GLS-1 inhibitor or a selective inhibitor of GLS-1.
  • the invention comprises a method of treating a subject in need of treatment by administering a glutaminase inhibitor to the subject, wherein the subject has: a deregulated NRF2/KEAP1 pathway; hyperactive NRF2 signaling; a loss-of-function mutation in the KEAP1 nucleic acid; a a gain-of- function mutation in NRF2 nucleic acid; or an increase in intracellular concentration of glutathione.
  • Figure 1 shows the inhibition of GLS by IACS-011393.
  • Figure 1 A shows the inhibition of GAC by varying concentrations of IACS-011393.
  • Figure IB shows the target engagement measured after treatment with IACS-011393.
  • Figure 1C shows that A549 cells utilize glutamine anaplerosis to drive respiration.
  • Figure ID shows the different sensitivities of different NSCLC lines to IACS-011393.
  • Figure 2 shows metabolic alterations in response to IACS-011393.
  • Figure 2A shows the results of the metabolic analysis of A549 and H727 cells treated with 1 ⁇ IACS-011393 for 24 hours.
  • Figure 2B shows glutaminase inhibition by IACS-011393 prevents the conversion of glutamine to glutathione.
  • Figure 2C shows a schematic representation of the incorporation of carbons from fully labeled 13C-Glutamine into glutathione in the absence or presence of the GLSi, IACS-011393.
  • Figure 3 shows that mutations in the Keapl/Nrf2 pathway, a major regulator of redox balance, predict sensitivity to GLSi in pre-clinical models of NSCLC.
  • Figure 3 A shows the differential sensitivity to IACS-011393 is predicted by Keapl/Nrf2 status.
  • Figure 3B shows that the mutations in responder cell lines fall within important functional domains of Keapl and Nrf2.
  • Figure 3C shows that target engagement is seen in both responder and non-responder cell lines, although responder cell lines are hyper- dependent on GLS-mediated glutaminolysis, while non-responder lines are less dependent on this process.
  • Figure 4 shows that GLS-dependence is driven by addiction to glutathione - mediated redox maintenance in Keapl /Nrf2 mutant cell lines.
  • Figure 4 A shows that glutathione levels are decreased after treatment with IACS-011393 in responder cell lines.
  • Figure 4B shows the loss of glutathione after treatment with a glutaminase inhibitor leads to an accumulation of intracellular reactive oxygen species(ROS).
  • Figure 4C shows that accumulation of DNA damage after IACS-011393 treatment can be rescued by application of cell-permeable glutathione.
  • Figure 4D shows that application of exogenous glutathione to responder cell lines rescues IACS-011393-induced proliferation defects.
  • Figure 5 shows that acquired resistance to IACS-011393 is derived when cells activate alternative mechanisms to maintain redox balance.
  • Figure 5A shows that cells treated chronically with GLSi acquire resistance to IACS-011393.
  • Figure 5B shows that clones resistant to GLSi display activation of alternative pathways to maintain redox balance.
  • Figure 5C shows that clones resistant to GLSi display altered transcriptional profiles to produce alternative sources of glutamate and reducing power.
  • Figure 5D shows a global overview of acquired adaptation to glutaminase inhibition.
  • the invention relates to the discovery of biomarkers and mechanisms associated with the responsiveness to treatment with glutaminase (GLS) inhibitors and methods to treat such patients.
  • GLS glutaminase
  • alterations to the NRF2/KEAP1 pathway sensitize patients to GLS inhibition.
  • KEAP1 a ubiquitin ligase adaptor protein, is a negative regulator of NRF2, the key transcription factor that activates the cellular antioxidant response.
  • NRF2/KEAP1 pathway an increase in the intracellular concentration of glutathione, or mutations in KEAP1 or NRF2 (for example, a loss-of- function mutation in KEAP1, or a gain-of-function mutation in NRF2) confer a dependence of tumors on reduced glutathione, the major endogenous antioxidant comprised of glycine, cysteine, and glutamine-derived glutamate. Inhibition of GLS reduces the steady state levels of glutathione, thus shifting the redox balance of tumor cells.
  • Nuclear factor (erythroid-derived 2)-like 2, also known as NRF2 is a transcription factor that in humans is encoded by the NFE2L2 gene. NRF2 is
  • NRF2 antioxidant response pathway is the primary cellular defense against the cytotoxic effects of oxidative stress. Among other effects, NRF2 increases the expression of several antioxidant enzymes. As NRF2 regulates a major cellular defense mechanism, tight regulation is crucial to maintain cellular homeostasis. Activation of this pathway is important in preventing human diseases, such as cancer, neurodegenerative disease, cardiovascular diseases, ischemia, diabetes, pulmonary fibrosis, and inflammatory diseases. Conversely, high constitutive levels of NRF2 occur in many tumors or cancer cell lines. Moreover, overexpression of NRF2 in cancer cells protects them from the cytotoxic effects of anticancer therapies, resulting in chemo- and/or radioresistance.
  • NRF2 Under normal or unstressed conditions, NRF2 is kept in the cytoplasm by a cluster of proteins that degrade it quickly. Under oxidative stress, NRF2 is not degraded, but instead travels to the nucleus where it binds to a DNA promoter and initiates transcription of antioxidative genes and their proteins. NRF2 is kept in the cytoplasm by Kelch like-ECH-associated protein 1 (KEAP1) and Cullin 3 which degrade NRF2 by ubiquitination. Cullin 3 ubiquitinates its substrate, NRF2. KEAP1 is a substrate adaptor, which helps Cullin 3 ubiquitinate NRF2.
  • NRF2 When NRF2 is ubiquitinated, it is transported to the proteasome, where it is degraded and its components recycled. Under normal conditions NRF2 has a half-life of only 20 minutes. Oxidative stress or electrophilic stress disrupts critical cysteine residues in KEAPl, disrupting the KEAPl -Cul3 ubiquitination system.
  • NRF2 When NRF2 is not ubiquitinated, it builds up in the cytoplasm, and translocates into the nucleus. In the nucleus, it combines (forms a heterodimer) with a small Maf protein and binds to the Antioxidant Response Element (ARE) in the upstream promoter region of many antioxidative genes, and initiates their transcription.
  • ARE Antioxidant Response Element
  • the NRF2/KEAP1 pathway is the major regulator of cytoprotective responses to endogenous and exogenous stresses caused by reactive oxygen species (ROS) and electrophiles.
  • the key signaling proteins within the pathway are the transcription factor NRF2 that binds together with small Maf proteins to the antioxidant response element (ARE) in the regulatory regions of target genes, and KEAPl, the repressor protein that binds to NRF2 and promotes its degradation by the ubiquitin proteasome pathway.
  • KEAPl is a very cysteine-rich protein, mouse KEAPl having a total of 25 and human 27 cysteine residues, most of which can be modified in vitro by different oxidants and electrophiles. Three of these residues, C151, C273 and C288, have been shown to play a functional role by altering the conformation of KEAPl leading to nuclear translocation of NRF2 and subsequent target gene expression.
  • the invention comprises a method of treating a subject having a disorder in need of treatment.
  • the method comprises (a) determining: (i) that the NRF2/KEAP1 pathway in said subject is deregulated, (ii) that NRF2 signaling in said subject is hyperactive, (iii) that the nucleic acid of said subject comprises a loss-of-function mutation in KEAPl, (iv) that the nucleic acid of said subject comprises a gain-of-function mutation in NRF2; or (v) an increase in the intracellular concentration of glutathione in said subject; and (b) administering a glutaminase inhibitor to the subject.
  • a method of treating a subject having a disorder in need of treatment comprising determining that the subject has an altered NRF2/KEAP1 pathway, and administering a glutaminase inhibitor to the subject.
  • the invention comprises a method of treating a subject having a disorder in need of treatment by administering a glutaminase inhibitor to the subject, wherein the subject has an altered NRF2/KEAP1 pathway.
  • the NRF2/KEAP1 pathway may be altered in one of several different mechanisms.
  • the NRF2/KEAP1 pathway in said subject is deregulated and ubiquitination of NRF2 which ultimately leads to its degradation is prevented.
  • the NRF2/KEAP1 pathway may be deregulated due to a mutation in either NRF2 or KEAPl .
  • the mutation may be any mutation that disrupts the function of KEAPl or a mutation in either KEAPl or NRF2 prevents the binding of KEAPl to NRF2.
  • the mutation is a somatic mutation.
  • the NRF2/KEAP1 pathway may be deregulated due to epigenetic silencing of KEAPl expression.
  • Epigenetic silencing of KEAPl expression results in constitutively activated NRF2 signaling.
  • hypermethylation at the promoter region of KEAPl results in epigenetic silencing of KEAPl expression.
  • Subjects with epigenetic silencing of KEAPl expression are good candidates for treatment with glutaminase inhibitor.
  • he NRF2/KEAP1 pathway may be deregulated due to accumulation of disruptor proteins leading to dissociation of the KEAPl -NRF2 complex.
  • disruptor proteins include, but are not limited to, cyclin- dependent kinase inhibitor, p21, and polyubiquitin binding protein, p62.
  • Transcriptional induction of NRF2 is another mechanism by which the NRF2/KEAP1 pathway would be deregulated.
  • the transcriptional induction of NRF2 may, for example, be caused by oncogenic signaling due to elements including, but not limited to, K-Ras, B-Raf and c-Myc.
  • the NRF2/KEAP1 pathway may be deregulated due to post-trans lational modification of KEAPl that affects binding of KEAPl to NRF2. Any of the amino acid residues of KEAPl may undergo post-translational modification.
  • the cysteine residues of KEAPl are modified. Post-translational modification of KEAPl, for example modification of the cysteine residues, includes, but is not limited to, succination of critical cysteine residues.
  • Critical KEAPl cysteine residues include, but are not limited to, those at positions 151, 273 and 288.
  • NRF2 signaling could be hyperactive due to several different factors, such as disruption of KEAPl -NRF2 binding or a reduction or loss in the function of KEAPl, caused, for example, by any of the mechanisms described above.
  • NRF2 signaling could also be hyperactive due to factors that affect expression of NRF2, including, but not limited to, induction of NRF2 by oncogenic signaling or mutations in NRF2 that result in increasing its expression or function.
  • the nucleic acid of the subject having a disorder in need of treatment comprises a mutation in one of the proteins involved in the
  • the mutation is a somatic mutation.
  • the nucleic acid encoding the KEAPl protein comprises a mutation, for example, a loss-of-function mutation, or a mutation that results in the reduction or the loss of the KEAPl protein's ability to bind NRF2.
  • the nucleic acid encoding the NRF2 protein comprises a mutation, for example, a gain-of-function mutation, or a mutation that results in the reduction or the loss of the NRF2 protein's ability to bind KEAPl or any other protein involved in the ubiquitination of NRF2.
  • a method of treating a subject having a disorder in need of treatment comprising determining that an increase in the intracellular concentration of glutathione in said subject and administering a glutaminase inhibitor to the subject.
  • concentration of glutathione (GSH) in a subject may be raised.
  • Non- limiting examples of such mechanisms include deregulated NRF2 activity or a deregulated NRF2/KEAP1 pathway as described above.
  • the intracellular concentration of glutathione in a subject is increased due to increased expression or activity of
  • glutathione-related enzymes include ⁇ - glutamylcysteine ligase and ⁇ -glutamyl-transpeptidase.
  • intracellular concentration of glutathione is increased due to increased expression or activity of amino acid transporters.
  • the invention comprises a method of treating a subject having a disorder in need of treatment by administering a glutaminase inhibitor to the subject, wherein the subject has: a deregulated NRF2/KEAP1 pathway; hyperactive NRF2 signaling; a loss-of- function mutation in the KEAPl nucleic acid; a gain-of- function mutation in NRF2 nucleic acid; or an increase in intracellular concentration of glutathione.
  • the subject has a deregulated NRF2/KEAP1 pathway.
  • the subject has hyperactive NRF2 signaling.
  • the subject has a loss-of-function mutation in the KEAPl nucleic acid.
  • the subject has a gain-of-function mutation in NRF2 nucleic acid.
  • the subject has an increase in intracellular concentration of glutathione.
  • the subject has more than one of the foregoing.
  • the invention comprises a method of treating a subject having a disorder in need of treatment by administering a compound that inhibits glutathione production to a subject.
  • the method comprises optionally obtaining a biological sample from a subject, determining that the NRF2/KEAP1 pathway in said subject is deregulated, or that NRF2 signaling in said subject is hyperactive, or determining the presence of a loss-of-function mutation in KEAPl or a gain-of-function mutation in NRF2 of said subject, or more than one of the foregoing, and administering a compound that inhibits glutathione production.
  • the compound inhibits amino acid transport or glutathione synthesis.
  • the compound that inhibits glutathione production is a glutaminase inhibitor.
  • the subject has a deregulated NRF2/KEAP1 pathway.
  • the subject has hyperactive NRF2 signaling.
  • the subject has a loss-of-function mutation in the KEAP1 nucleic acid.
  • the subject has a gain-of-function mutation in NRF2 nucleic acid.
  • the subject has an increase in intracellular concentration of glutathione. In certain embodiments, the subject has more than one of the foregoing.
  • the invention comprises a method of evaluating whether to administer a compound that inhibits glutathione production to a subject.
  • the method comprises optionally obtaining a biological sample from a subject, determining that the NRF2/KEAP1 pathway in said subject is deregulated, or that NRF2 signaling in said subject is hyperactive, or determining the presence of a loss-of-function mutation in KEAP1 or a gain-of-function mutation in NRF2 of said subject and determining that the compound that inhibits glutathione production should be
  • the compound inhibits amino acid transport or glutathione synthesis.
  • the subject has a deregulated NRF2/KEAP1 pathway.
  • the subject has hyperactive NRF2 signaling.
  • the subject has a loss-of-function mutation in the KEAP1 nucleic acid.
  • the subject has a gain-of-function mutation in NRF2 nucleic acid.
  • the subject has an increase in intracellular concentration of glutathione. In certain embodiments, the subject has more than one of the foregoing.
  • a glutaminase inhibitor or a compound that inhibits glutathione production for use as a medicament in the treatment of a disorder in need of treatment, in a subject in which (i) the NRF2/KEAP1 pathway in said subject is deregulated, (ii) NRF2 signaling in said subject is hyperactive, (iii) that the nucleic acid of said subject comprises a loss-of-function mutation in KEAP1, (iv) the nucleic acid of said subject comprises a gain-of-function mutation in NRF2; or (v) the subject has an increase in the intracellular concentration of glutathione.
  • a glutaminase inhibitor or a compound that inhibits glutathione production for treatment of a disorder in need of treatment in a subject in which (i) the NRF2/KEAP1 pathway in said subject is deregulated, (ii) NRF2 signaling in said subject is hyperactive, (iii) that the nucleic acid of said subject comprises a loss- of-function mutation in KEAP1, (iv) the nucleic acid of said subject comprises a gain-of- function mutation in NRF2; or (v) the subject has an increase in the intracellular concentration of glutathione.
  • a glutaminase inhibitor or a compound that inhibits glutathione production for use in the manufacture of a medicament for the treatment of a disorder in need of treatment, in a subject in which (i) the NRF2/KEAP1 pathway in said subject is deregulated, (ii) NRF2 signaling in said subject is hyperactive, (iii) that the nucleic acid of said subject comprises a loss-of- function mutation in KEAP1, (iv) the nucleic acid of said subject comprises a gain-of-function mutation in NRF2; or (v) the subject has an increase in the intracellular concentration of glutathione.
  • the NRF2/KEAP1 pathway in said subject is deregulated because of:
  • the NRF2/KEAP1 pathway in said subject is deregulated because of a somatic mutation in KEAPl or the KEAPl binding domain of NRF2. In certain embodiments, the NRF2/KEAP1 pathway in said subject is deregulated because of DNA hypermethylation at the promoter region of KEAPl . In certain embodiments, the NRF2/KEAP1 pathway in said subject is deregulated because of accumulation of cyclin-dependent kinase inhibitor p21 or polyubiquitin binding protein p62. In certain embodiments, the NRF2/KEAP1 pathway in said subject is deregulated because of transcriptional induction of NRF2 by K-Ras, B-Raf or c-Myc oncogenic signaling.
  • the NRF2/KEAP1 pathway in said subject is deregulated because of post-translational modification of KEAP1 cysteine residues. In certain embodiments, the NRF2/KEAP1 pathway in said subject is deregulated because of expression of a microRNA. that down-regulates EAP1 levels.
  • the subject has an increase in the intracellular concentration of glutathione (GSH), which is caused by:
  • the increase in the intracellular concentration of glutathione (GSH) in said subject is caused by deregulated NRF2 activity. In certain embodiments, the increase in the intracellular concentration of glutathione (GSH) in said subject is caused by increased expression of GSH-related enzymes. In certain embodiments, the increase in the intracellular concentration of glutathione (GSH) in said subject is caused by increased expression or activity of amino acid transporters.
  • the disorder is a cancer.
  • the cancer is one or a variant of: bladder cancer, bone marrow cancer, breast cancer, cancer of the central nervous system, cervical cancer, colon cancer, endometrial cancer, cancer of the gastric system, head and neck cancer, kidney cancer, liver cancer, lung cancer, muscle cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, or thyroid cancer.
  • the cancer is lung cancer.
  • the cancer is non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the glutaminase inhibitor is a selective inhibitor of GLS-1.
  • the GLS-1 inhibitor binds an allosteric pocket on the solvent exposed region of the GLS-1 dimer in the binding pocket present in the vicinity of Leu321, Phe322, Leu323, and Tyr394 from both monomers.
  • the glutaminase inhibitor is chosen from the list of compounds provided in Table 1 below.
  • the compound is chosen from any combination of the compounds provided in Table 1 , or a salt or polymorph thereof.
  • the compound is chosen from any two, three, four, five, six, seven, eight, none or ten of the compounds provided in Table 1 , or a salt or polymorph thereof.
  • the subject is human.
  • methods disclosed herein further comprise administering another pharmaceutically active compound.
  • the disorder is a cancer and the other pharmaceutically active compound is an anti-cancer agent.
  • the anti-cancer agent is a chosen from a platinum-based agent, a taxane-based agent, an immunotherapy, a targeted therapy.
  • the targeted therapy is an inhibitor of MEK kinase, HSP90, CDK4, or the mTOR pathway.
  • methods disclosed herein further comprise administering non-chemical methods of cancer treatment.
  • the method further comprises administering radiation therapy.
  • the method further comprises administering surgery, thermoablation, focused ultrasound therapy, cryotherapy, or any combination thereof.
  • methods disclosed herein administer the active agent (e.g., a compound that inhibits glutathione production, glutaminase inhibitor, or selective GLS-1 inhibitor) as a pharmaceutical composition comprising a pharmaceutically acceptable excipient or carrier.
  • the pharmaceutical composition is formulated for oral administration.
  • pharmaceutical composition is formulated as a tablet or capsule.
  • the pharmaceutical composition is formulated for parenteral administration.
  • any embodiment disclosed herein, and particularly any embodiment disclosed above in paragraphs [021] - [051] above or in paragraphs [0148] - [0182] below, may be combined with any one or more of these embodiments to form a new compound or class of compounds, or pharmaceutical composition comprising it, or method of use employing it, provided the combination is not mutually exclusive.
  • a combination embodiment wherein the subject is human and the disorder in need of treatment is cancer is valid because the recited limitations are not mutually exclusive.
  • the term "and/or" when used in a list of two or more items, means that any one of the listed items can be employed by itself or in combination with any one or more of the listed items.
  • the expression “A and/or B” is intended to mean either or both of A and B, i.e., A alone, B alone or A and B in combination.
  • the expression “A, B and/or C” is intended to mean A alone, B alone, C alone, A and B in combination, A and C in combination, B and C in combination or A, B, and C in combination.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
  • combination therapy means the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • altered NRF2/KEAP 1 pathway or a “NRF2/KEAP 1 pathway that is altered” are used interchangeably herein and refer to a subject or a cell or a cell line in which any of the following are present: (i) a deregulated NRF2/KEAP1 pathway;
  • GLS-1 inhibitor is used herein to refer to a compound that exhibits an ICso with respect to GLS-1 activity of no more than about 100 ⁇ and more typically not more than about 50 ⁇ , as measured in the GLS-1 enzyme assay described generally herein below.
  • ICso is that concentration of inhibitor that reduces the activity of an enzyme (e.g., GLS-1) to half-maximal level. Certain compounds disclosed herein have been discovered to exhibit inhibition against GLS-1.
  • compounds will exhibit an ICso with respect to GLS-1 of no more than about 10 ⁇ ; in further embodiments, compounds will exhibit an ICso with respect to GLS-1 of no more than about 5 ⁇ ; in yet further embodiments, compounds will exhibit an ICso with respect to GLS-1 of not more than about 1 ⁇ ; in yet further embodiments, compounds will exhibit an ICso with respect to GLS-1 of not more than about 200 nM, as measured in the GLS-1 enzymatic assay described herein.
  • inhibitor selective for GLS-1 or a “selective inhibitor of GLS-1” are used interchangeably herein and refer to inhibitors that are about 100 times more selective for GLS-1 than for GLS-2 as measured in any assay known to one of skill in the art that measures the activity of the enzyme.
  • An example of such an assay includes, but is not limited to, the GLS-1 enzyme assay (GLS-1 Enzymatic Activity Assay) described below.
  • terapéuticaally acceptable refers to those compounds (or salts, prodrugs, tautomers, zwitterionic forms, etc.) which are suitable for use in contact with the tissues of patients without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • treatment of a patient is intended to include prevention, prophylaxis, attenuation, amelioration and therapy. Treatment may also include prevention of disease. Prevention of a disease may involve complete protection from disease, for example as in the case of prevention of infection with a pathogen, or may involve prevention of disease progression. For example, prevention of a disease may not mean complete foreclosure of any effect related to the diseases at any level, but instead may mean prevention of the symptoms of a disease to a clinically significant or detectable level. Prevention of diseases may also mean prevention of progression of a disease to a later stage of the disease.
  • subject or "patient” are used interchangeably herein to mean all mammals including humans. Examples of subjects include, but are not limited to, humans, monkeys, dogs, cats, horses, cows, goats, sheep, pigs, and rabbits. In one embodiment, the patient is a human.
  • acyl refers to a carbonyl attached to an alkenyl, alkyl, aryl, cycloalkyl, heteroaryl, heterocycle, or any other moiety were the atom attached to the carbonyl is carbon.
  • An “acetyl” group refers to
  • alkylcarbonyl or “alkanoyl” group refers to an alkyl group attached to the parent molecular moiety through a carbonyl group. Examples of such groups include methylcarbonyl and ethylcarbonyl. Examples of acyl groups include formyl, alkanoyl and aroyl.
  • alkenyl refers to a straight-chain or branched-chain hydrocarbon radical having one or more double bonds and containing from 2 to 20 carbon atoms. In certain embodiments, the alkenyl will comprise from 2 to 6 carbon atoms.
  • alkoxy refers to an alkyl ether radical, wherein the term alkyl is as defined below.
  • suitable alkyl ether radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec- butoxy, tert-butoxy, and the like.
  • alkyl refers to a straight- chain or branched-chain alkyl radical containing from 1 to 20 carbon atoms. In certain embodiments, the alkyl will comprise from 1 to 10 carbon atoms. In further embodiments, the alkyl will comprise from 1 to 10 carbon atoms.
  • the alkyl will comprise from 1 to 6 carbon atoms.
  • Alkyl groups may be optionally substituted as defined herein.
  • alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, hexyl, octyl, noyl and the like.
  • alkylene as used herein, alone or in combination, refers to a saturated aliphatic group derived from a straight or branched chain saturated hydrocarbon attached at two or more positions, such as methylene (-CH2-). Unless otherwise specified, the term “alkyl” may include “alkylene” groups.
  • alkylamino refers to an alkyl group attached to the parent molecular moiety through an amino group. Suitable alkylamino groups may be mono- or dialkylated, forming groups such as, for example, N- methylamino, N-ethylamino, N,N-dimethylamino, ⁇ , ⁇ -ethylmethylamino and the like.
  • alkylidene refers to an alkenyl group in which one carbon atom of the carbon-carbon double bond belongs to the moiety to which the alkenyl group is attached.
  • alkylthio refers to an alkyl thioether (R-S-) radical wherein the term alkyl is as defined above and wherein the sulfur may be singly or doubly oxidized.
  • suitable alkyl thioether radicals include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, iso-butylthio, sec- butylthio, tert-butylthio, methanesulfonyl, ethanesulfmyl, and the like.
  • alkynyl refers to a straight-chain or branched chain hydrocarbon radical having one or more triple bonds and containing from 2 to 20 carbon atoms. In certain embodiments, the alkynyl comprises from 2 to 6 carbon atoms. In further embodiments, the alkynyl comprises from 2 to 4 carbon atoms.
  • alkynylene refers to a carbon-carbon triple bond attached at two positions such as ethynylene (-C:::C-, -C ⁇ C-).
  • alkynyl radicals include ethynyl, propynyl, hydroxypropynyl, butyn-l-yl, butyn-2-yl, pentyn-l-yl, 3-methylbutyn- 1-yl, hexyn-2-yl, and the like.
  • alkynyl may include "alkynylene” groups.
  • acylamino as used herein, alone or in combination, embraces an acyl group attached to the parent moiety through an amino group.
  • An example of an “acylamino” group is acetylamino (CH3C(0)NH-).
  • amino refers to— NRR', wherein R and R' are independently selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted. Additionally, R and R' may combine to form heterocycloalkyl, either of which may be optionally substituted.
  • aryl as used herein, alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such polycyclic ring systems are fused together.
  • aryl embraces aromatic groups such as phenyl, naphthyl, anthracenyl, and phenanthryl.
  • combination refers to an aryl group attached to the parent molecular moiety through an alkenyl group.
  • arylalkoxy or “aralkoxy,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkoxy group.
  • arylalkyl or “aralkyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkyl group.
  • combination refers to an aryl group attached to the parent molecular moiety through an alkynyl group.
  • arylalkanoyl or “aralkanoyl” or “aroyl,” as used herein, alone or in combination, refers to an acyl radical derived from an aryl-substituted alkanecarboxylic acid such as benzoyl, napthoyl, phenylacetyl, 3-phenylpropionyl (hydrocinnamoyl), 4- phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like.
  • aryloxy refers to an aryl group attached to the parent molecular moiety through an oxy.
  • carbamate refers to an ester of carbamic acid (-NHCOO-) which may be attached to the parent molecular moiety from either the nitrogen or acid end, and which may be optionally substituted as defined herein.
  • O-carbamyl as used herein, alone or in combination, refers to a -OC(0)NRR', group with R and R' as defined herein.
  • N-carbamyl as used herein, alone or in combination, refers to a ROC(0)NR'- group, with R and R' as defined herein.
  • carbonyl when alone includes formyl [-C(0)H] and in combination is a -C(O)- group.
  • carboxyl or “carboxy,” as used herein, refers to -C(0)OH or the corresponding “carboxylate” anion, such as is in a carboxylic acid salt.
  • An "O-carboxy” group refers to a RC(0)0- group, where R is as defined herein.
  • a “C-carboxy” group refers to a -C(0)OR groups where R is as defined herein.
  • cyano as used herein, alone or in combination, refers to -CN.
  • cycloalkyl or, alternatively, “carbocycle,” as used herein, alone or in combination, refers to a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl group wherein each cyclic moiety contains from 3 to 12 carbon atom ring members and which may optionally be a benzo fused ring system which is optionally substituted as defined herein.
  • the cycloalkyl will comprise from 5 to 7 carbon atoms. Examples of such cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydronapthyl, indanyl,
  • bicyclic and tricyclic as used herein are intended to include both fused ring systems, such as decahydronaphthalene, octahydronaphthalene as well as the multicyclic (multicentered) saturated or partially unsaturated type.
  • the latter type of isomer is exemplified in general by, bicyclo[l,l,l]pentane, camphor, adamantane, and bicyclo[3,2,l]octane.
  • esters refers to a carboxy group bridging two moieties linked at carbon atoms.
  • ether refers to an oxy group bridging two moieties linked at carbon atoms.
  • halo or halogen
  • haloalkoxy refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • haloalkyl refers to an alkyl radical having the meaning as defined above wherein one or more hydrogens are replaced with a halogen. Specifically embraced are monohaloalkyl, dihaloalkyl and polyhaloalkyl radicals.
  • a monohaloalkyl radical for one example, may have an iodo, bromo, chloro or fluoro atom within the radical.
  • Dihalo and polyhaloalkyl radicals may have two or more of the same halo atoms or a combination of different halo radicals.
  • haloalkyl radicals include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
  • Haloalkylene refers to a haloalkyl group attached at two or more positions. Examples include fluoromethylene
  • heteroalkyl refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. Up to two heteroatoms may be consecutive, such as, for example, -CH2-NH-OCH3.
  • heteroaryl refers to a 3 to 15 membered unsaturated heteromonocyclic ring, or a fused monocyclic, bicyclic, or tricyclic ring system in which at least one of the fused rings is aromatic, which contains at least one atom selected from the group consisting of O, S, and N.
  • the heteroaryl will comprise from 5 to 7 carbon atoms.
  • heterocyclic rings are fused with aryl rings, wherein heteroaryl rings are fused with other heteroaryl rings, wherein heteroaryl rings are fused with heterocycloalkyl rings, or wherein heteroaryl rings are fused with cycloalkyl rings.
  • heteroaryl groups include pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, pyranyl, furyl, thienyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isothiazolyl, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, indazolyl, benzotriazolyl, benzodioxolyl, benzopyranyl, benzoxazolyl, benzoxadiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, benzothienyl, chromonyl,
  • Exemplary tricyclic heterocyclic groups include carbazolyl, benzidolyl, phenanthrolinyl, dibenzofuranyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • heterocycloalkyl and, interchangeably, “heterocycle,” as used herein, alone or in combination, each refer to a saturated, partially unsaturated, or fully unsaturated monocyclic, bicyclic, or tricyclic heterocyclic group containing at least one heteroatom as a ring member, wherein each the heteroatom may be independently selected from the group consisting of nitrogen, oxygen, and sulfur In certain
  • the hetercycloalkyl will comprise from 1 to 4 heteroatoms as ring members. In further embodiments, the hetercycloalkyl will comprise from 1 to 2 heteroatoms as ring members. In certain embodiments, the hetercycloalkyl will comprise from 3 to 8 ring members in each ring. In further embodiments, the hetercycloalkyl will comprise from 3 to 7 ring members in each ring. In yet further embodiments, the hetercycloalkyl will comprise from 5 to 6 ring members in each ring.
  • Heterocycloalkyl and “heterocycle” are intended to include sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems; additionally, both terms also include systems where a heterocycle ring is fused to an aryl group, as defined herein, or an additional heterocycle group.
  • heterocycle groups include aziridinyl, azetidinyl, 1,3-benzodioxolyl, dihydroisoindolyl,
  • heterocycle groups may be optionally substituted unless specifically prohibited.
  • hydrazinyl as used herein, alone or in combination, refers to two amino groups joined by a single bond, i.e., -N-N-.
  • hydroxyalkyl refers to a hydroxy group attached to the parent molecular moiety through an alkyl group.
  • isocyanato refers to a -NCO group.
  • isothiocyanato refers to a -NCS group.
  • linear chain of atoms refers to the longest straight chain of atoms independently selected from carbon, nitrogen, oxygen and sulfur.
  • lower means containing from 1 to and including 6 carbon atoms.
  • lower aryl means phenyl or naphthyl, either of which may be optionally substituted as provided.
  • lower heteroaryl means either 1) monocyclic heteroaryl comprising five or six ring members, of which between one and four the members may be heteroatoms selected from the group consisting of O, S, and N, or 2) bicyclic heteroaryl, wherein each of the fused rings comprises five or six ring members, comprising between them one to four heteroatoms selected from the group consisting of O, S, and N.
  • lower cycloalkyl as used herein, alone or in combination, means a monocyclic cycloalkyl having between three and six ring members. Lower cycloalkyls may be unsaturated. Examples of lower cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • lower heterocycloalkyl as used herein, alone or in combination, means a monocyclic heterocycloalkyl having between three and six ring members, of which between one and four may be heteroatoms selected from the group consisting of O, S, and N.
  • lower heterocycloalkyls include pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, and morpholinyl.
  • Lower heterocycloalkyls may be unsaturated.
  • lower amino refers to— NR ', wherein R and R' are independently selected from the group consisting of hydrogen, lower alkyl, and lower heteroalkyl, any of which may be optionally
  • R and R' of a lower amino group may combine to form a five- or six-membered heterocycloalkyl, either of which may be optionally substituted.
  • mercaptyl as used herein, alone or in combination, refers to an RS- group, where R is as defined herein.
  • perhaloalkoxy refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms.
  • perhaloalkyl refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
  • sulfonate refers the -SO3H group and its anion as the sulfonic acid is used in salt formation.
  • thia and thio refer to a -S- group or an ether wherein the oxygen is replaced with sulfur.
  • the oxidized derivatives of the thio group namely sulfmyl and sulfonyl, are included in the definition of thia and thio.
  • thiol as used herein, alone or in combination, refers to an -SH group.
  • thioformyl -C(S)H and in combination is a -C(S)- group.
  • N-thiocarbamyl refers to an ROC(S)NR'- group, with R and R' as defined herein.
  • O-thiocarbamyl refers to a -OC(S)NRR' , group with R and R' as defined herein.
  • thiocyanato refers to a -CNS group.
  • trihalomethanesulfonamido refers to a X 3 CS(0) 2 NR- group with X is a halogen and R as defined herein.
  • trihalomethanesulfonyl refers to a X 3 CS(0) 2 - group where X is a halogen.
  • trimethoxy refers to a X 3 CO- group where X is a halogen.
  • trimethysilyl as used herein, alone or in combination, refers to a silicone group substituted at its three free valences with groups as listed herein under the definition of substituted amino. Examples include trimethysilyl, tert- butyldimethylsilyl, triphenylsilyl and the like.
  • any definition herein may be used in combination with any other definition to describe a composite structural group.
  • the trailing element of any such definition is that which attaches to the parent moiety.
  • the composite group alkylamido would represent an alkyl group attached to the parent molecule through an amido group
  • the term alkoxyalkyl would represent an alkoxy group attached to the parent molecule through an alkyl group.
  • lower alkyl lower alkenyl, lower alkynyl, lower alkanoyl, lower heteroalkyl, lower heterocycloalkyl, lower haloalkyl, lower haloalkenyl, lower haloalkynyl, lower perhaloalkyl, lower perhaloalkoxy, lower cycloalkyl, phenyl, aryl, aryloxy, lower alkoxy, lower haloalkoxy, oxo, lower acyloxy, carbonyl, carboxyl, lower alkylcarbonyl, lower carboxyester, lower carboxamido, cyano, hydrogen, halogen, hydroxy, amino, lower alkylamino, arylamino, amido, nitro, thiol, lower alkylthio, lower haloalkylthio, lower perhaloalkylthio, arylthio, sulfonate
  • Two substituents may be joined together to form a fused five-, six-, or seven-membered carbocyclic or heterocyclic ring consisting of zero to three heteroatoms, for example forming methylenedioxy or ethylenedioxy.
  • An optionally substituted group may be unsubstituted (e.g., -CH2CH3), fully substituted (e.g., -CF2CF3), monosubstituted (e.g., -CH2CH2F) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g., -CH2CF3).
  • substituents are recited without qualification as to substitution, both substituted and unsubstituted forms are
  • R or the term R' appearing by itself and without a number designation, unless otherwise defined, refers to a moiety selected from the group consisting of hydrogen, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl and
  • heterocycloalkyl any of which may be optionally substituted.
  • stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art.
  • Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art.
  • the compounds disclosed herein may exist as geometric isomers. The present disclosure includes all cis, trans, syn, anti,
  • compounds may exist as tautomers; all tautomeric isomers are provided by this disclosure. Additionally, the compounds disclosed herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms.
  • bond refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered part of larger substructure.
  • a bond may be single, double, or triple unless otherwise specified.
  • a dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
  • the compounds disclosed herein can exist as therapeutically acceptable salts.
  • the present disclosure includes compounds listed above in the form of salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non-pharmaceutically acceptable salts may be of utility in the preparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable.
  • Pharmaceutical Salts Properties, Selection, and Use (Stahl, P. Heinrich. Wiley- VCHA, Zurich, Switzerland, 2002).
  • terapéuticaally acceptable salt represents salts or zwitterionic forms of the compounds disclosed herein which are water or oil-soluble or dispersible and therapeutically acceptable as defined herein.
  • the salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid.
  • Representative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, formate, fumarate, gentisate, glutarate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide,
  • hydroiodide 2-hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL- mandelate, mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, 2- naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproprionate, phosphonate, picrate, pivalate, propionate, pyroglutamate, succinate, sulfonate, tartrate, L-tartrate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, para- toluenesulfonate (p-tosylate), and undecanoate.
  • basic groups in the compounds disclosed herein can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides.
  • acids which can be employed to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Salts can also be formed by coordination of the compounds with an alkali metal or alkaline earth ion.
  • the present disclosure contemplates sodium, potassium, magnesium, and calcium salts of the compounds disclosed herein, and the like.
  • Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxy group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • the cations of therapeutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium,
  • tetraethylammonium methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, ⁇ , ⁇ -dimethylaniline, N- methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, ⁇ , ⁇ -dibenzylphenethylamine, 1-ephenamine, and ⁇ , ⁇ '-dibenzylethylenediamine.
  • Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.
  • a salt of a compound can be made by reacting the appropriate compound in the form of the free base with the appropriate acid.
  • the compound is a compound that inhibits glutathione production or activity. In certain embodiments, the compound inhibits amino acid or glutathione transport. In some embodiments, the compound is a glutaminase inhibitor.
  • the compound is a GLS-1 inhibitor, for example, a selective inhibitor of GLS-1. It is known that GLS-1 forms a tetramer (PNAS 2012, 109, 7705).
  • the GLS-1 inhibitor occupies an allosteric pocket on the solvent exposed region between two GLS-1 dimers.
  • the GLS-1 inhibitor may, for example, bind GLS-1 in an allosteric pocket on the solvent exposed region of the GLS-1 dimer in the binding pocket present in the vicinity of amino acids Leu 321, Phe322, Leu323, and Tyr394 from both monomers.
  • the inventors propose that key interactions are made within a hydrophobic cluster that comprises Leu321, Phe322, Leu323, and Tyr394 from both monomers which forms the allosteric pocket. Binding of the glutaminase inhibitor, for example, a GLS-1 inhibitor, induces a dramatic conformational change near the catalytic site rendering the enzyme inactive.
  • the compound (and its molecular mass) is provided in Table 1 below, or a salt or polymorph thereof.
  • the compound is chosen from any combination of the compounds provided in Table 1 below, or a salt or polymorph thereof.
  • the compound is chosen from any two, three, four, five, six, seven, eight, none or ten of the compounds provided in Table 1 below, or a salt or polymorph thereof.
  • the compound is (S)-2-hydroxy-2-phenyl-N-(5-(4-(6- (2-(3 -(trifluoromethoxy)pheny l)acetamido)pyridazin-3 -yl)butyl)- 1 ,3 ,4-thiadiazol-2- yl)acetamide, or a salt or polymorph thereof.
  • the compound is N,N * -(5,5 * -(2,2 * -thiobis(ethane-2,l-diyl))bis(l,3,4-thiadiazole-5,2-diyl))bis(2- phenylacetamide), or a salt or polymorph thereof.
  • the compound is (S)-2-hydroxy-2-(pyridin-2-yl)-N-(5-(4-(6-(2-(3-
  • the compound is N,N * -(5,5 * -(2,2 , -sulfonylbis(ethane-2,l-diyl))bis(l,3,4-thiadiazole-5,2-diyl))bis(2- (pyridin-2-yl)acetamide), or a salt or polymorph thereof.
  • the compound is N-methyl- 1 - ⁇ 4-[6-(2- ⁇ 4-[3-(trifluoromethoxy)phenyl]pyridin-2- yl ⁇ acetamido)pyridazin-3-yl]butyl ⁇ -lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is l-(2-fluoro-4-(5-(2- (pyridin-2-yl)acetamido)- 1 ,3 ,4-thiadiazol-2-yl)butyl)-N-((4-(trifluoromethyl)pyridin-2- yl)methyl)-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is l-(2-fluoro-4-(6-(2-(4-(trifluoromethyl)pyridin-2- yl)acetamido)pyridazin-3-yl)butyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is N-(pyridin-2-ylmethyl)- 5 -(3 -(6-(2-(3 -(trifluoromethoxy)phenyl)acetamido)pyridazin-3 -yl)pyrrolidin- 1 -yl)- 1 ,3 ,4- thiadiazole-2-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)- 1 -(2-fluoro-4-(6-(2-(4-(3-(trifluoromethoxy)phenyl)pyridin-2- yl)acetamido)pyridazin-3-yl)butyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)-l-(2-fluoro-4-(6-(2- (4-(trifluoromethyl)pyridin-2-yl)acetamido)pyridazin-3-yl)butyl)-N-methyl-lH- 1,2,3- triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)- 1 -(2-fluoro-4-(6-(2-(6-methyl-4-(trifluoromethyl)pyridin-2- yl)acetamido)pyridazin-3-yl)butyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)-l-(4-(6-(2-(4- (cyclopropyldifluoromethyl)pyridin-2-yl)acetamido)pyridazin-3-yl)-2-fluorobutyl)-N- methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)-l-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)pyridin-2- yl)acetamido)pyridazin-3-yl)-2-fluorobutyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)-l-(2-fluoro- 4-(6-(2-(l-(3-(trifluoromethoxy)phenyl)-lH-imidazol-4-yl)acetamido)pyridazin-3- yl)butyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is l-(4-(6-(2-(4-cyclobutoxypyridin-2- yl)acetamido)pyridazin-3-yl)butyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is l-(4-(6-(2-(4- cyclobutoxypyridin-2-yl)acetamido)pyridazin-3-yl)-2-fluorobutyl)-N-methyl-lH- 1,2,3- triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is l-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)pyridin-2-yl)acetamido)pyridazin-3- yl)butyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is l-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)pyridin-2- yl)acetamido)pyridazin-3-yl)-2-fluorobutyl)-N-methyl-lH-l,2,3-triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is (R)-l-(4-(6-(2- (4-cyclopropylpyridin-2-yl)acetamido)pyridazin-3-yl)-2-fluorobutyl)-N-methyl-lH- 1,2,3- triazole-4-carboxamide, or a salt or polymorph thereof.
  • the compound is 5-(3-(6-(2-(pyridin-2-yl)acetamido)pyridazin-3-yl)pyrrolidin-l-yl)-N-((4- (trifluoromethyl)pyridin-2-yl)methyl)-l,3,4-thiadiazole-2-carboxamide, or a salt or polymorph thereof.
  • the compound is N,N'-(5,5'-(cyclohexane- l,3-diyl)bis(l,3,4-thiadiazole-5,2-diyl))bis(2-phenylacetamide), or a salt or polymorph thereof.
  • the compound is N,N'-(5,5'-((lS,3S)-cyclohexane-l,3- diyl)bis(l,3,4-thiadiazole-5,2-diyl))bis(2-phenylacetamide), or a salt or polymorph thereof.
  • the compound is N,N'-(5,5'-((lR,3R)-cyclohexane-l,3- diyl)bis(l,3,4-thiadiazole-5,2-diyl))bis(2-phenylacetamide), or a salt or polymorph thereof.
  • the GLS-1 inhibitor is disclosed in United States Application No. 14/791,284 filed July 3, 2015.
  • the GLS-1 inhibitor is of Formula I:
  • n is chosen from 3, 4, and 5;
  • each R x and R y is independently chosen from alkyl, cyano, H, and halo, wherein two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and A 2 are independently chosen from C-H, C-F, and N;
  • R 1 and R 4 are independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(R 3 ) 2 C(0)R 3 , C(R 3 ) 2 C(0)N(R 3 ) 2 , C(R 3 ) 2 N(R 3 ) 2 , C(R 3 ) 2 NR 3 C(0)R 3 , C(R 3 ) 2 NR 3 C(0)OR 3 , C(R 3 ) 2 NR 3 C(0)N(R 3 ) 2 , C(R 3 ) 2 NR 3 S(0)R 3 , C(R 3 ) 2 NR 3 S(0) 2 R 3 , N(R 3 ) 2 , NR 3 C(0)R 3 , NR 3 C
  • each R 1 and R 4 may be optionally substituted with between 0 and 3 R z groups;
  • R 2 is chosen from alkyl, heterocycloalkyl, cyano, cycloalkyl, H, halo, and haloalkyl, wherein R 1 and R 2 together with the atoms to which they are attached optionally form an form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R 3 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 3 may be optionally substituted with between 0 and 3 R z groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl, haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • each R 6 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein two R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R x groups; and
  • Z is heteroaryl, which may be optionally substituted.
  • the GLS-1 inhibitor is of la:
  • n is chosen from 3, 4, and 5;
  • each R x and R y is independently chosen from alkyl, cyano, H, and halo, wherein two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and A 2 are independently chosen from C-H, C-F, and N;
  • Z 1 is chosen from C and N;
  • Z 2 , Z 3 , and Z 4 are independently chosen from N, O, S, and CH, wherein at least one of Z 1 , Z 2 , Z 3 , and Z 4 is chosen from N, O, and S;
  • R 1 and R 4 are independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(R 3 ) 2 C(0)R 3 , C(R 3 ) 2 C(0)N(R 3 ) 2 , C(R 3 ) 2 N(R 3 ) 2 , C(R 3 ) 2 NR 3 C(0)R 3 , C(R 3 ) 2 NR 3 C(0)OR 3 , C(R 3 ) 2 NR 3 C(0)N(R 3 ) 2 ,
  • each R 1 and R 4 may be optionally substituted with between 0 and 3 R z groups;
  • R 2 is chosen from alkyl, heterocycloalkyl, cyano, cycloalkyl, H, halo, and haloalkyl, wherein R 1 and R 2 together with the atoms to which they are attached optionally form an form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R 3 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 3 may be optionally substituted with between 0 and 3 R z groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxomen N(R 6 ) 2 , NR 6 C(0)C(R 6 )3,
  • each R 6 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein two R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R x groups.
  • the GLS-1 inhibitor is of formula lb:
  • n is chosen from 3, 4, and 5;
  • each R x and R y is independently chosen from alkyl, cyano, H, and halo, wherein two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and A 2 are independently chosen from C-H, C-F, and N;
  • Z 1 is chosen from C and N;
  • Z 2 is chosen from N, CH, and C(O);
  • Z 3 , and Z 4 are independently chosen from N and CH, wherein at least one of Z 1 , Z 2 , Z 3 , and Z 4 is N;
  • R 1 and R 4 are independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(R 3 ) 2 C(0)R 3 , C(R 3 ) 2 C(0)N(R 3 ) 2 , C(R 3 ) 2 N(R 3 ) 2 , C(R 3 ) 2 NR 3 C(0)R 3 , C(R 3 ) 2 NR 3 C(0)OR 3 , C(R 3 ) 2 NR 3 C(0)N(R 3 ) 2 ,
  • each R 1 and R 4 may be optionally substituted with between 0 and 3 R z groups;
  • R 2 and R 5 are chosen from alkyl, heterocycloalkyl, cyano, cycloalkyl, H, halo, and haloalkyl, wherein R 1 and R 2 together with the atoms to which they are attached optionally form an form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups, wherein R 4 and R 5 together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R 3 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 3 may be optionally substituted with between 0 and 3 R z groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheteroarylalkyl, haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl, haloalkylcycloalkylalkylalkyl, haloalkylheteroaryl, haloalkylheteroarylalkyl, haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl, halocycloalkylalkyl, hal
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 6 ) 2 , NR 6 C(0)C(R 6 )3,
  • each R 6 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein two R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R x groups.
  • the GLS-1 inhibitor is disclosed in United States Application 14/791,206, filed July 2, 2015.
  • the GLS-1 inhibitor is of Formula II:
  • n is chosen from 3, 4, and 5;
  • each R x and R y is independently chosen from alkyl, cyano, H, and halo, wherein two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and A 2 are independently chosen from N and CH;
  • a 3 is chosen from N and CR 2 ;
  • R 1 is chosen from alkenyl, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, C(0)N(R 3 ) 2 , and C(0)C(R 3 ) 3 , wherein R 1 may be optionally substituted with between 0 and 3 R z groups;
  • R 2 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(0)N(R 3 ) 2 , C(0)C(R 3 ) 3 , C(0)OH, C(0)OC(R 3 ) 3 , wherein R 1 and R 2 together with the atoms to which they are attached optionally form an form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R 3 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 3 may be optionally substituted with between 0 and 3 R z groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • R 4 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, N(R 3 ) 2 , NR 3 C(0)C(R 3 ) 3 , NR 3 C(0)OC(R 3 ) 3 ,
  • R 4 may be optionally substituted with between 0 and 3 R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, oxo, N(R 6 ) 2 , NR 6 C(0)C(R 6 ) 3 , NR 6 C(0)OC(R 6 ) 3 , NR 6 C(0)N(R 6 ) 2 , NR 6 S(0)C(R 6 ) 3 , NR 6 S(0) 2 C(R 6 ) 3 , C(0)N(R 6 ) 2 , S(0)N(R 6 ) 2 , S(0) 2 N(R 6 ) 2 , C(0)C(R 6 ) 3 , SC(R 6 ) 3 , S(0)C(R 6 ) 3 , and S(0) 2 C(R
  • each R 6 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein two R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R x groups; and
  • Z is heteroaryl, which may be optionally substituted.
  • the GLS-1 inhibitor is of formula lib:
  • n is chosen from 3, 4, and 5;
  • each R x and R y is independently chosen from alkyl, cyano, H, and halo, wherein two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and A 2 are independently chosen from N and CH;
  • a 3 is chosen from N and CR 2 ;
  • Z 1 is chosen from C and N;
  • Z 2 , Z 3 , and Z 4 are independently chosen from N, O, S, and CH, wherein at least one of Z 1 , Z 2 , Z 3 , and Z 4 is chosen from N, O, and S;
  • R 1 is chosen from alkenyl, alkyl, aryl, arylalkyl, cyano, cycloalkyl,
  • cycloalkylalkyl H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, C(0)N(R 3 ) 2 , and C(0)C(R 3 )3, wherein R 1 may be optionally substituted with between 0 and 3 R z groups;
  • R 2 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(0)N(R 3 ) 2 , C(0)C(R 3 ) 3 , C(0)OH, C(0)OC(R 3 ) 3 , wherein R 1 and R 2 together with the atoms to which they are attached optionally form an form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups; each R 3 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H,
  • R 4 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, N(R 3 ) 2 , NR 3 C(0)C(R 3 ) 3 , NR 3 C(0)OC(R 3 ) 3 ,
  • R 4 may be optionally substituted with between 0 and 3 R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, oxo, N(R 6 ) 2 , NR 6 C(0)C(R 6 ) 3 , NR 6 C(0)OC(R 6 ) 3 , NR 6 C(0)N(R 6 ) 2 , NR 6 S(0)C(R 6 ) 3 , NR 6 S(0) 2 C(R 6 ) 3 , C(0)N(R 6 ) 2 , S(0)N(R 6 ) 2 , S(0) 2 N(R 6 ) 2 , C(0)C(R 6 ) 3 , SC(R 6 ) 3 , S(0)C(R 6 ) 3 , and S(0) 2 C(R
  • each R 6 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein two R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R x groups.
  • the GLS-1 inhibitor is of formula lib:
  • n is chosen from 3, 4, and 5;
  • each R x and R y is independently chosen from alkyl, cyano, H, and halo, wherein two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and A 2 are independently chosen from N and CH;
  • a 3 is chosen from N and CR 2 ;
  • Z 1 is chosen from C and N;
  • Z 2 is chosen from N, CH, and C(O);
  • Z 3 , and Z 4 are independently chosen from N and CH, wherein at least one of Z 1 , Z 2 , Z 3 , and Z 4 is N;
  • R 1 is chosen from alkenyl, alkyl, aryl, arylalkyl, cyano, cycloalkyl,
  • cycloalkylalkyl H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, C(0)N(R 3 ) 2 , and C(0)C(R 3 )3, wherein R 1 may be optionally substituted with between 0 and 3 R z groups;
  • R 2 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(0)N(R 3 ) 2 , C(0)C(R 3 ) 3 , C(0)OH, C(0)OC(R 3 ) 3 , wherein R 1 and R 2 together with the atoms to which they are attached optionally form an form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • each R 3 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 3 may be optionally substituted with between 0 and 3 R z groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R z groups;
  • R 4 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, N(R 3 ) 2 , NR 3 C(0)C(R 3 ) 3 , NR 3 C(0)OC(R 3 ) 3 , NR 3 C(0)N(R 3 ) 2 , NR 3 S(0)C(R 3 ) 3 , NR 3 S(0) 2 C(R 3 ) 3 , C(0)N(R 3 ) 2 , S(0)N(R 3 ) 2 ,
  • R 4 may be optionally substituted with between 0 and 3 R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, oxo, N(R 6 ) 2 , NR 6 C(0)C(R 6 ) 3 , NR 6 C(0)OC(R 6 ) 3 , NR 6 C(0)N(R 6 ) 2 , NR 6 S(0)C(R 6 ) 3 , NR 6 S(0) 2 C(R 6 ) 3 , C(0)N(R 6 ) 2 , S(0)N(R 6 ) 2 , S(0) 2 N(R 6 ) 2 , C(0)C(R 6 ) 3 , SC(R 6 ) 3 , S(0)C(R 6 ) 3 , and S(0) 2 C(R
  • each R 6 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein two R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with between 0 and 3 R x groups.
  • the GLS-1 inhibitor is disclosed in 14/791,186, filed July 2, 2015.
  • the GLS-1 inhibitor is of Formula III:
  • n is chosen from 3, 4, and 5;
  • each R x and R Y is independently chosen from alkyl, cyano, H, and halo, or two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 is chosen from C and N;
  • a 2 , A 3 , and A 4 are independently chosen from N, O, S, and CH, wherein at least one of A 1 , A 2 , A 3 , and A 4 is chosen from N, O, and S;
  • R 1 and R 2 are each independently chosen from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein R 1 and R 2 each may be optionally substituted with one to three R z groups, wherein R 1 and R 2 together with the atoms to which they are attached optionally form an heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups;
  • R 3 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(R 4 ) 2 C(0)R 4 , C(R 4 ) 2 C(0)N(R 4 ) 2 , C(R 4 ) 2 N(R 4 ) 2 , C(R 4 ) 2 NR 4 C(0)R 4 , C(R 4 ) 2 NR 4 C(0)OR 4 , C(R 4 ) 2 NR 4 C(0)N(R 4 ) 2 , C(R 4 ) 2 NR 4 S(0)R 4 , C(R 4 ) 2 NR 4 S(0) 2 R 4 , N(R 4 ) 2 , NR 4 C(0)R 4 , NR 4 C(0)OR 4
  • each R 4 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 4 may be optionally substituted with one to three R z groups, wherein two R 4 groups together with the atoms to which they are attached optionally form an heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl, halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 ,
  • NR 5 C(0)OR 5 NR 5 C(0)N(R 5 ) 2 , NR 5 S(0)R 5 , NR 5 S(0) 2 R 5 , C(0)N(R 5 ) 2 , S(0)N(R 5 ) 2 , S(0) 2 N(R 5 ) 2 , C(0)R 5 , C(0)OR 5 , SR 5 , S(0)R 5 , and S(0) 2 R 5 ;
  • each R 5 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein two R 5 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups; and
  • Z is a monocyclic heteroaryl, which may be optionally substituted.
  • the GLS-1 inhibitor is of Formula Ilia:
  • n is chosen from 3, 4, and 5;
  • each R x and R Y is independently chosen from alkyl, cyano, H, and halo, or two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 and Z 1 are independently chosen from C and N;
  • a 2 , A 3 , A 4 , Z 2 , Z 3 , and Z 4 are independently chosen from N, O, S, and CH, wherein at least one of A 1 , A 2 , A 3 , and A 4 and at least one of Z 1 , Z 2 , Z 3 , and Z 4 is chosen from N, O, and S;
  • R 1 and R 2 are each independently chosen from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl,
  • R 1 and R 2 each may be optionally substituted with one to three R z groups, wherein R 1 and R 2 together with the atoms to which they are attached optionally form an heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups;
  • R 3 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(R 4 ) 2 C(0)R 4 , C(R 4 ) 2 C(0)N(R 4 ) 2 , C(R 4 ) 2 N(R 4 ) 2 , C(R 4 ) 2 NR 4 C(0)R 4 , C(R 4 ) 2 NR 4 C(0)OR 4 , C(R 4 ) 2 NR 4 C(0)N(R 4 ) 2 , C(R 4 ) 2 NR 4 S(0)R 4 , C(R 4 ) 2 NR 4 S(0) 2 R 4 , N(R 4 ) 2 , NR 4 C(0)R 4 , NR 4 C(0)OR 4
  • each R 4 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 4 may be optionally substituted with one to three R z groups, wherein two R 4 groups together with the atoms to which they are attached optionally form an heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 ,
  • NR 5 C(0)OR 5 NR 5 C(0)N(R 5 ) 2 , NR 5 S(0)R 5 , NR 5 S(0) 2 R 5 , C(0)N(R 5 ) 2 , S(0)N(R 5 ) 2 , S(0) 2 N(R 5 ) 2 , C(0)R 5 , C(0)OR 5 , SR 5 , S(0)R 5 , and S(0) 2 R 5 ;
  • each R 5 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein two R 5 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups.
  • the GLS-1 inhibitor is of Formula Illb:
  • n is chosen from 3, 4, and 5;
  • each R x and R Y is independently chosen from alkyl, cyano, H, and halo, or two R x groups together with the atoms to which they are attached optionally form a cycloalkyl ring;
  • a 1 is chosen from C and N;
  • a 2 , A 3 , and A 4 are independently chosen from N, O, S, and CH, wherein at least one of A 1 , A 2 , A 3 , and A 4 is chosen from N, O, and S;
  • Z 1 is C
  • Z 2 Z 3 and Z 4 are independently chosen from N and CH, wherein at least one of Z 1 , Z 2 , Z 3 , and Z 4 is N;
  • R 1 and R 2 are each independently chosen from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl,
  • R 1 and R 2 each may be optionally substituted with one to three R z groups, wherein R 1 and R 2 together with the atoms to which they are attached optionally form an heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups;
  • R 3 is chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, hydroxyl, C(R 4 ) 2 C(0)R 4 , C(R 4 ) 2 C(0)N(R 4 ) 2 , C(R 4 ) 2 N(R 4 ) 2 , C(R 4 ) 2 NR 4 C(0)R 4 , C(R 4 ) 2 NR 4 C(0)OR 4 , C(R 4 ) 2 NR 4 C(0)N(R 4 ) 2 , C(R 4 ) 2 NR 4 S(0)R 4 , C(R 4 ) 2 NR 4 S(0) 2 R 4 , N(R 4 ) 2 , NR 4 C(0)R 4 , NR 4 C(0)OR 4
  • each R 4 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein each R 4 may be optionally substituted with one to three R z groups, wherein two R 4 groups together with the atoms to which they are attached optionally form an heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 ,
  • NR 5 C(0)OR 5 NR 5 C(0)N(R 5 ) 2 , NR 5 S(0)R 5 , NR 5 S(0) 2 R 5 , C(0)N(R 5 ) 2 , S(0)N(R 5 ) 2 , S(0) 2 N(R 5 ) 2 , C(0)R 5 , C(0)OR 5 , SR 5 , S(0)R 5 , and S(0) 2 R 5 ;
  • each R 5 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein two R 5 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups; and
  • R 6 is chosen from, alkyl, cyano, cycloalkyl, H, halo, haloalkyl, and
  • heterocycloalkyl wherein R 3 and R 6 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R z groups.
  • the GLS-1 inhibitor is of Formula IIIc:
  • R x is chosen from fluoro and H;
  • R 1 is chosen from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein R 1 may be optionally substituted with one to three R z groups;
  • each R 4 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein R 4 may be optionally substituted with one to three R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • each R 5 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein two R 5 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups.
  • the GLS-1 inhibitor is of Formula IIIc-1 :
  • R x is chosen from fluoro and H
  • R 1 is chosen from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein R 1 may be optionally substituted with one to three R z groups;
  • each R 4 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein R 4 may be optionally substituted with one to three R z groups;
  • each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • each R 5 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl,
  • heterocycloalkyl, and heterocycloalkylalkyl, wherein two R 5 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups.
  • the GLS-1 inhibitor is of Formula IIIc-2:
  • R x is chosen from fluoro and H
  • R 1 is chosen from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein R 1 may be optionally substituted with one to three R z groups;
  • each R 4 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, heterocycloalkylalkyl, and hydroxyl, wherein R 4 may be optionally substituted with one to three R z groups; each R z group is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • each R 5 is independently chosen from alkenyl, alkoxy, alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl,
  • heterocycloalkyl, and heterocycloalkylalkyl, wherein two R 5 groups together with the atoms to which they are attached optionally form an aryl, cycloalkyl, heteroaryl, or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups.
  • the GLS-1 inhibitor is of Formula Hid:
  • R x is chosen from fluoro and H
  • each of R Z1 and R Z2 is independently chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, and oxo.
  • the GLS-1 inhibitor is of Formula Hie:
  • R x is chosen from fluoro and H
  • R Z1 is chosen from alkenyl, alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl, alkoxyheterocycloalkylalkyl, alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl, alkylcycloalkylalkyl, alkylheteroaryl, alkylheteroarylalkyl, alkylheterocycloalkyl, alkylheterocycloalkylalkyl, aryl, arylalkyl, arylalkyloxy, arylhaloalkyl, aryloxy, cyano, cycloalkyl, cycloalkylalkyl
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, and oxo.
  • the GLS-1 inhibitor is one as disclosed in United States provisional Application No. 62/187,160 filed June 30, 2015. [0171] In certain embodime -1 inhibitor is of Formula IV:
  • n is chosen from 1 and 2;
  • R 1 is chosen from NR 3 C(0)R 3 , NR 3 C(0)OR 3 , NR 3 C(0)N(R 3 ) 2 , C(0)N(R 3 ) 2 , and
  • each R 3 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl,
  • each R 3 may be optionally substituted with one to three R x groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an heteroaryl or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups;
  • R 2 is chosen from NR 4 C(0)R 4 , NR 4 C(0)OR 4 , NR 4 C(0)N(R 4 ) 2 , C(0)N(R 4 ) 2 and
  • each R 4 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 4 may be optionally substituted with one to three R x groups, wherein two R 4 groups together with the atoms to which they are attached optionally form an heteroaryl or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups;
  • each R x group is independently chosen from alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl, alkoxyheteroaryl, alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkyl, haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl, haloalkoxyheteroarylalkyl, haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 , NR 5 C(0)OR 5 , NR 5 C(0)N(R 5 ) 2 , C(0)N(R 5 ) 2 , and C(0)R 5 ;
  • each R 5 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, which may be optionally substituted with one to three R z groups;
  • R z is chosen from alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl;
  • A is a monocyclic heteroaryl, which may be optionally substituted with one to three R z groups;
  • Z is a monocyclic heteroaryl, which may be optionally substituted with one to three R z groups.
  • the GLS-1 inhibitor is of Formula IVa:
  • n is chosen from 1 and 2;
  • R 1 is chosen from NR 3 C(0)R 3 , NR 3 C(0)OR 3 , NR 3 C(0)N(R 3 ) 2 , C(0)N(R 3 ) 2 , and
  • each R 3 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl,
  • each R 3 may be optionally substituted with one to three R x groups, wherein two R 3 groups together with the atoms to which they are attached optionally form an heteroaryl or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups;
  • R 2 is chosen from NR 4 C(0)R 4 , NR 4 C(0)OR 4 , NR 4 C(0)N(R 4 ) 2 , C(0)N(R 4 ) 2 and
  • each R 4 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 4 may be optionally substituted with one to three R x groups, wherein two R 4 groups together with the atoms to which they are attached optionally form an heteroaryl or heterocycloalkyl ring, which may be optionally substituted with one to three R x groups;
  • each R x group is independently chosen from alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl,
  • alkoxyheteroaryl alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 , NR 5 C(0)OR 5 , NR 5 C(0)N(R 5 ) 2 , C(0)N(R 5 ) 2 , and C(0)R 5 ;
  • each R 5 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, which may be optionally substituted with one to three R z groups; and
  • R z is chosen from alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl.
  • the GLS-1 inhibitor is of Formula IVb:
  • n is chosen from 1 and 2;
  • R 3 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 3 may be optionally substituted with one to three R x groups;
  • R 4 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 4 may be optionally substituted with one to three R x groups;
  • each R x group is independently chosen from alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl,
  • alkoxyheteroaryl alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 , NR 5 C(0)OR 5 , NR 5 C(0)N(R 5 ) 2 , C(0)N(R 5 ) 2 , and C(0)R 5 ;
  • each R 5 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, which may be optionally substituted with one to three R z groups; and
  • R z is chosen from alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl.
  • the GLS-1 inhibitor is of Formula IVc:
  • n is chosen from 1 and 2;
  • R 3 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 3 may be optionally substituted with one to three R x groups;
  • R 4 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 4 may be optionally substituted with one to three R x groups;
  • each R x group is independently chosen from alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl,
  • alkoxyheteroaryl alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 , NR 5 C(0)OR 5 , NR 5 C(0)N(R 5 ) 2 , C(0)N(R 5 ) 2 , and C(0)R 5 ;
  • each R 5 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, which may be optionally substituted with one to three R z groups; and
  • R z is chosen from alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl.
  • the GLS-1 inhibitor is of Formula IVd:
  • n is chosen from 1 and 2;
  • R 3 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 3 may be optionally substituted with one to three R x groups;
  • R 4 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 4 may be optionally substituted with one to three R x groups;
  • each R x group is independently chosen from alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl,
  • alkoxyheteroaryl alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl,
  • haloalkoxyheteroarylalkyl haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 , NR 5 C(0)OR 5 , NR 5 C(0)N(R 5 ) 2 , C(0)N(R 5 ) 2 , and C(0)R 5 ;
  • each R 5 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, which may be optionally substituted with one to three R z groups; and
  • R z is chosen from alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl.
  • the GLS-1 inhibitor is of Formula IVe:
  • n is chosen from 1 and 2;
  • R 3 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 3 may be optionally substituted with one to three R x groups;
  • R 4 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, wherein each R 4 may be optionally substituted with one to three R x groups;
  • each R x group is independently chosen from alkoxy, alkoxyalkyl, alkoxyaryl, alkoxyarylalkyl, alkoxycycloalkyl, alkoxycycloalkylalkyl, alkoxyhaloalkyl,
  • alkoxyheteroaryl alkoxyheteroarylalkyl, alkoxyheterocycloalkyl,
  • alkoxyheterocycloalkylalkyl alkyl, alkylaryl, alkylarylalkyl, alkylcycloalkyl,
  • haloalkoxycycloalkyl haloalkoxycycloalkyl, haloalkoxycycloalkylalkyl, haloalkoxyheteroaryl, haloalkoxyheteroarylalkyl, haloalkoxyheterocycloalkyl, haloalkoxyheterocycloalkylalkyl, haloalkyl, haloalkylaryl, haloalkylarylalkyl, haloalkylcycloalkyl,
  • haloalkylcycloalkylalkyl haloalkylheteroaryl, haloalkylheteroarylalkyl,
  • haloalkylheterocycloalkyl haloalkylheterocycloalkyl, haloalkylheterocycloalkylalkyl, haloaryl, haloarylalkyl, haloarylalkyloxy, haloaryloxy, halocycloalkyl, halocycloalkylalkyl,
  • halocycloalkylalkyloxy halocycloalkylalkyloxy, halocycloalkyloxy, haloheteroaryl, haloheteroarylalkyl, haloheteroarylalkyloxy, haloheteroaryloxy, haloheterocycloalkyl,
  • haloheterocycloalkylalkyl haloheterocycloalkylalkyl, haloheterocycloalkylalkyloxy, haloheterocycloalkyloxy, heteroaryl, heteroarylalkyl, heteroarylalkyloxy, heteroarylhaloalkyl, heteroaryloxy, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkyloxy,
  • heterocycloalkylhaloalkyl heterocycloalkyloxy, hydroxyl, oxo, N(R 5 ) 2 , NR 5 C(0)R 5 , NR 5 C(0)OR 5 , NR 5 C(0)N(R 5 ) 2 , C(0)N(R 5 ) 2 , and C(0)R 5 ;
  • each R 5 is independently chosen from alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, H, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl, which may be optionally substituted with one to three R z groups; and
  • R z is chosen from alkyl, aryl, arylalkyl, cyano, cycloalkyl, cycloalkylalkyl, H, halo, haloalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, and heterocycloalkylalkyl.
  • the GLS-1 inhibitor is one as disclosed in
  • the GLS-1 inhibitor is of formula Al,
  • Y independently for each occurrence, represents H or CH 2 0(CO)R7;
  • R7 independently for each occurrence, represents H or substituted or
  • alkyl unsubstituted alkyl, alkoxy, aminoalkyl, alkylaminoalkyl, heterocyclylalkyl, arylalkyl, or heterocyclylalkoxy;
  • Z represents H or R3(CO);
  • Ri and R 2 each independently represent H, alkyl, alkoxy or hydroxy
  • R3 independently for each occurrence, represents substituted or unsubstituted alkyl, hydroxyalkyl, aminoalkyl, acylaminoalkyl, alkenyl, alkoxy, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl,
  • heterocyclylalkyl heteroaryl, heteroarylalkyl, heteroaryloxy, heteroaryloxyalkyl or C(R8)(R9)(Rio), N(R 4 )(R5) or OR 6 , wherein any free hydroxyl group may be acylated to form C(0)Rv;
  • R 4 and R5 each independently represent H or substituted or unsubstituted alkyl, hydroxyalkyl, acyl, aminoalkyl, acylaminoalkyl, alkenyl, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, wherein any free hydroxyl group may be acylated to form C(0)R7;
  • R 6 independently for each occurrence, represents substituted or unsubstituted alkyl, hydroxyalkyl, aminoalkyl, acylaminoalkyl, alkenyl, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, wherein any free hydroxyl group may be acylated to form C(0)R7; and
  • R8, R9 and Rio each independently represent H or substituted or unsubstituted alkyl, hydroxy, hydroxyalkyl, amino, acylamino, aminoalkyl, acylaminoalkyl, alkoxycarbonyl, alkoxycarbonylamino, alkenyl, alkoxy, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, or Rs and R9 together with the carbon to which they are attached, form a carbocyclic or heterocyclic ring system, wherein any free hydroxyl group may be acylated to form C(0)R7, and wherein at least two of Rs, R and Rio are not H.
  • the GLS-1 inhibitor is one as disclosed in
  • the GLS-1 inhibitor is of formula A2,
  • Y independently for each occurrence, represents H or CH20(CO)R7;
  • R7 independently for each occurrence, represents H or substituted or
  • alkyl unsubstituted alkyl, alkoxy, aminoalkyl, alkylaminoalkyl, heterocyclylalkyl, arylalkyl, or heterocyclylalkoxy;
  • Z represents H or R3(CO);
  • Ri and R2 each independently represent H, alkyl, alkoxy or hydroxy
  • R3 represents substituted or unsubstituted alkyl, hydroxyalkyl, aminoalkyl, acylaminoalkyl, alkenyl, alkoxy, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, heteroaryloxyalkyl or C(Rs)(R9)(Rio), N(R 4 )(R5) or OR 6 , wherein any free hydroxyl group may be acylated to form C(0)R7;
  • R 4 and R5 each independently for each occurrence represent H or substituted or unsubstituted alkyl, hydroxyalkyl, acyl, aminoalkyl, acylaminoalkyl, alkenyl, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or
  • heteroaryloxyalkyl wherein any free hydroxyl group may be acylated to form C(0)R7;
  • R 6 represents substituted or unsubstituted alkyl, hydroxyalkyl, aminoalkyl, acylaminoalkyl, alkenyl, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl,
  • heteroaryloxy, or heteroaryloxyalkyl, wherein any free hydroxyl group may be acylated to form C(0)Rv;
  • R8, R and Rio each independently for each occurrence represent H or substituted or unsubstituted alkyl, hydroxy, hydroxyalkyl, amino, acylamino, aminoalkyl, acylaminoalkyl, alkoxycarbonyl, alkoxycarbonylamino, alkenyl, alkoxy, alkoxyalkyl, aryl, arylalkyl, aryloxy, aryloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, or R8 and R9 together with the carbon to which they are attached, form a carbocyclic or heterocyclic ring system, wherein any free hydroxyl group may be acylated to form C(0)R7, and wherein at least two of Rs, R9 and Rio are not H;
  • R11 represents aryl, arylalkyl, aryloxy, aryloxyalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, wherein the aryl or heteroaryl ring is substituted with either -OCHF2 or -OCF3 and is optionally further substituted, or R11 represents C(Ri2)(Ri3)(Ri4), N(R 4 )(Ri4) or ORi 4 , wherein any free hydroxyl group may be acylated to form C(0)Rv;
  • R12 and Ri3 each independently respresent H or substituted or unsubstituted alkyl, hydroxy, hydroxyalkyl, amino, acylamino, aminoalkyl, acylaminoalkyl, alkoxycarbonyl, alkoxycarbonylamino, alkenyl, alkoxy, alkoxyalkyl, aryl, arylalkyl, aryloxy,
  • aryloxyalkyl cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, wherein any free hydroxyl group may be acylated to form C(0)R7, and wherein both of R12 and R13 are not H;
  • Ri4 represents aryl, arylalkyl, aryloxy, aryloxyalkyl, heteroaryl, heteroarylalkyl, heteroaryloxy, or heteroaryloxyalkyl, wherein the aryl or heteroaryl ring is substituted with either -OCHF2 or -OCF3 and is optionally further substituted.
  • the GLS-1 inhibitor is one as disclosed in
  • the GLS-1 inhibitor is of formula A3,
  • X is C3-C7 cycloalkylene
  • each Ri and R 2 is independently -NH 2 , -N(R3)-C(0)-R 4 , -C(0)-N(R 3 )-R4, N(R 3 )- C(0)-0-R 4 , -N(R3)-C(0)-N(R 3 )-R4 or -N(R3)-C(0)-SR 4 ;
  • each R 3 is independently hydrogen, Ci-6 alkyl or aryl
  • each R4 is independently Ci-6 alkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, cycloalkylalkyl, heterocyclylalkyl, or heterocyclyl, each of which is substituted with 0-3 occurrences of R5;
  • each R5 is independently Ci-6 alkyl, Ci-6 alkoxy, Ci-6 thioalkoxy, Ci-6 haloalkyl, C3-7 cycloalkyl, C3-7 cycloalkylalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl,
  • heterocyclylalkyl heterocyclyl, heterocyclyl, cyano, halo, oxo, -OH, -OCF 3 , -SO2-C1-6 alkyl, -NO2, - N(R7)-C(0)- Ci-6 alkyl, -N(R7) 2 , or two adjacent R5 moieties, taken together with the atoms to which they are attached form a heterocyclyl;
  • each R 6 is independently hydrogen, fluoro, Ci-6 alkyl, -OH, -NH 2 , -NH(CH 3 ), N(CH 3 ) 2 , or Ci-6 alkoxy;
  • each R7 is independently hydrogen or Ci-6 alkyl
  • n 0, 1, or 2;
  • n 0, 1, or 2;
  • o 1, 2 or 3;
  • p 1, 2 or 3.
  • the GLS-1 inhibitor is one as disclosed in
  • the GLS-1 inhibitor is of formula A4,
  • each Ri and R2 is independently Ci-e alkylene-R 4 , -N(R 3 )-R 4 , -N(R 3 )-C(0)-R 4 , C(0)-N(R 3 )-R 4 , -N(R 3 )-C(0)-0-R 4 , -N(R 3 )-C(0)-N(R 3 )-R 4 , -0-C(0)-N(R 3 )-R 4 , - N(R 3 )C(0)-Ci-6 alkylene-C(0)-R 4 , -N(R 3 )-C(0)-Ci- 6 alkylene-N(R 3 )-C(0)-R 4 or C(0)CH2-N(R 3 )-C(0)-R 4 ;
  • each R 3 and R 3a is independently hydrogen, Ci-6 alkyl or aryl
  • each R 4 is independently Ci-6 alkyl, Ci-6 alkenyl, aryl, heteroaryl, aralkyl, heteroaralkyl, heterocyclylalkyl, heterocyclyl, cycloalkyl or cycloalkylalkyl, each of which is substituted with 0-3 occurrences of R5, or two adjacent R5 moieties, taken together with the atoms to which they are attached form a heterocyclyl, heteroaryl, cycloalkyl or aryl;
  • each R 6 is independently hydrogen, fluoro, OH or Ci-6 alkyl
  • each R7 is independently hydrogen, Ci-6 alkyl, -OH, -SH, cyano, halo, -CF 3 ,OCF 3 , -SO2-C1-6 alkyl, -NO2, -N(R 7 )-C(0)-Ci-6 alkyl, -N(Re) 2 or Ci-e alkoxy;
  • p 1, 2 or 3.
  • compositions which comprise one or more of certain compounds disclosed herein, or one or more
  • compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee- making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound of the subject disclosure or a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof ("active ingredient”) with the carrier which constitutes one or more accessory ingredients.
  • active ingredient a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • the compounds of the present invention may be administered orally, including swallowing, so the compound enters the gastrointestinal tract, or is absorbed into the blood stream directly from the mouth, including sublingual or buccal administration.
  • compositions for oral administration include solid formulations such as tablets, pills, cachets, lozenges and hard or soft capsules, which can contain liquids, gels, powders, or granules.
  • the amount of drug present may be from about 0.05% to about 95% by weight, more typically from about 2% to about 50% by weight of the dosage form.
  • tablets or capsules may contain a disintegrant, comprising from about 0.5%) to about 35% by weight, more typically from about 2% to about 25% of the dosage form.
  • disintegrants include methyl cellulose, sodium or calcium carboxymethyl cellulose, croscarmellose sodium, polyvinylpyrrolidone, hydroxypropyl cellulose, starch and the like.
  • Suitable binders for use in a tablet, include gelatin, polyethylene glycol, sugars, gums, starch, hydroxypropyl cellulose and the like.
  • Suitable diluents, for use in a tablet include mannitol, xylitol, lactose, dextrose, sucrose, sorbitol and starch.
  • Suitable surface active agents and glidants for use in a tablet or capsule, may be present in amounts from about 0.1% to about 3% by weight, and include polysorbate 80, sodium dodecyl sulfate, talc and silicon dioxide.
  • Suitable lubricants for use in a tablet or capsule, may be present in amounts from about 0.1% to about 5% by weight, and include calcium, zinc or magnesium stearate, sodium stearyl fumarate and the like.
  • Tablets may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with a liquid diluent. Dyes or pigments may be added to tablets for identification or to characterize different combinations of active compound doses.
  • Liquid formulations can include emulsions, solutions, syrups, elixirs and suspensions, which can be used in soft or hard capsules.
  • Such formulations may include a pharmaceutically acceptable carrier, for example, water, ethanol, polyethylene glycol, cellulose, or an oil.
  • the formulation may also include one or more emulsifying agents and/or suspending agents.
  • compositions for oral administration may be formulated as immediate or modified release, including delayed or sustained release, optionally with enteric coating.
  • a pharmaceutical composition comprises a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • Compounds of the present invention may be administered directly into the blood stream, muscle, or internal organs by injection, e.g., by bolus injection or continuous infusion.
  • Suitable means for parenteral administration include intravenous, intra-muscular, subcutaneous intraarterial, intraperitoneal, intrathecal, intracranial, and the like.
  • Suitable devices for parenteral administration include injectors (including needle and needle-free injectors) and infusion methods.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials.
  • parenteral formulations are aqueous solutions containing excipients, including salts, buffering, suspending, stabilizing and/or dispersing agents, antioxidants, bacteriostats, preservatives, and solutes which render the formulation isotonic with the blood of the intended recipient, and carbohydrates.
  • Parenteral formulations may also be prepared in a dehydrated form (e.g., by lyophilization) or as sterile non-aqueous solutions. These formulations can be used with a suitable vehicle, such as sterile water. Solubility-enhancing agents may also be used in preparation of parenteral solutions.
  • compositions for parenteral administration may be formulated as immediate or modified release, including delayed or sustained release.
  • Compounds may also be formulated as depot preparations. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • Compounds of the present invention may be administered topically (for example to the skin, mucous membranes, ear, nose, or eye) or transdermally.
  • Formulations for topical administration can include, but are not limited to, lotions, solutions, creams, gels, hydrogels, ointments, foams, implants, patches and the like.
  • Carriers that are pharmaceutically acceptable for topical administration formulations can include water, alcohol, mineral oil, glycerin, polyethylene glycol and the like.
  • Topical administration can also be performed by, for example, electroporation, iontophoresis, phonophoresis and the like.
  • the active ingredient for topical administration may comprise from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10%> w/w; less than 5% w/w; from 2% w/w to 5% w/w; or from 0.1% to 1% w/w of the formulation.
  • Compositions for topical administration may be formulated as immediate or modified release, including delayed or sustained release.
  • Suppositories for rectal administration of the compounds of the present invention can be prepared by mixing the active agent with a suitable non-irritating excipient such as cocoa butter, synthetic mono-, di-, or triglycerides, fatty acids, or polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature, and which will therefore melt in the rectum and release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic mono-, di-, or triglycerides, fatty acids, or polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature, and which will therefore melt in the rectum and release the drug.
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner.
  • Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
  • compounds may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray or powder.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds according to the disclosure may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures.
  • Preferred unit dosage formulations are those containing an effective dose, as herein recited, or an appropriate fraction thereof, of the active ingredient.
  • the precise amount of compound administered to a patient will be the responsibility of the attendant physician.
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of
  • administration route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the indication or condition being treated.
  • route of administration may vary depending on the condition and its severity.
  • the methods of the invention can be used to treat any subject in need of treatment.
  • subjects or patients include, but are not limited to, humans, monkeys, deer, camel, pets and companion animals, including, but not limited to, dogs, cats, horses, rabbits, and guinea pigs; livestock, including, but not limited to, cows, buffalo, bison, mules, goats, sheep and pigs.
  • livestock including, but not limited to, cows, buffalo, bison, mules, goats, sheep and pigs.
  • the subject is a human.
  • the methods of the invention provide for the administration of a glutaminase inhibitor or a compound that inhibits glutathione production for the treatment of several diseases and disorders characterized by an altered NRF2/KEAP1 pathway.
  • the disorder is a cancer.
  • provided herein is a method of treatment of a disease or disorder comprising the administration of a glutaminase inhibitor or a compound that inhibits glutathione production.
  • diseases or disorders include, but are not limited to, cancers.
  • a glutaminase inhibitor or a compound that inhibits glutathione production for use as a medicament is provided herein.
  • provided herein is the use of a glutaminase inhibitor or a compound that inhibits glutathione production for use in the manufacture of a medicament.
  • a glutaminase inhibitor or a compound that inhibits glutathione production for use in the manufacture of a medicament is provided herein.
  • medicament for the treatment of a disease or disorder including, but not limited to, the treatment of various cancers.
  • the cancer is one or a variant of: bladder cancer, breast cancer, bone marrow cancer, cancer of the central nervous system, cervical cancer, colon cancer, endometrial cancer, cancer of the gastric system, head and neck cancer, kidney cancer, liver cancer, lung cancer, muscle cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, or thyroid cancer.
  • the cancer is a lung cancer, including, but not limited to, non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • methods described herein are used to treat a disease condition comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or pharmaceutically acceptable salt thereof, wherein the condition is cancer which has developed resistance to chemotherapeutic drugs and/or ionizing radiation.
  • the compounds of the present invention can be used, alone or in combination with other pharmaceutically active compounds, to treat conditions such as those disclosed hereinabove.
  • the compound(s) of the present invention and other pharmaceutically active compound(s) can be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially.
  • the present invention comprises methods for treating a condition by administering to the subject a therapeutically-effective amount of one or more compounds of the present invention and one or more additional pharmaceutically active compounds.
  • a glutaminase inhibitor or a compound that inhibits glutathione production for use as a medicament in combination with one or more additional pharmaceutically active compounds.
  • a glutaminase inhibitor or a compound that inhibits glutathione production and one or more additional pharmaceutically active compounds for the manufacturing of a medicament.
  • a glutaminase inhibitor or a compound that inhibits glutathione production and one or more additional pharmaceutically active compounds for the manufacturing of a medicament for the treatment of a disease or disorder, including, but not limited to, the treatment of various cancers.
  • composition comprising one or more compounds of the present invention, one or more additional pharmaceutically active compounds, and a pharmaceutically acceptable carrier.
  • the one or more additional pharmaceutically active compounds is chosen from anti-cancer drugs, anti-proliferative drugs, and antiinflammatory drugs.
  • the anti-cancer agent is chosen from a platinum-based agent, a taxane-based agent, an immunotherapy, and a targeted therapy.
  • the targeted therapy is an inhibitor of MEK kinase, HSP90, CDK4, or the mTOR pathway.
  • Glutaminase inhibitors e.g., GLS-1 inhibitors, disclosed herein are also optionally used in combination with other therapeutic reagents that are selected for their therapeutic value for the condition to be treated.
  • the compounds described herein and, in embodiments where combination therapy is employed, other agents do not have to be administered in the same pharmaceutical composition and, because of different physical and chemical characteristics, are optionally administered by different routes.
  • the initial administration is generally made according to established protocols and then, based upon the observed effects, the dosage, modes of administration and times of administration subsequently modified. In certain instances, it is appropriate to administer a glutaminase inhibitor compound, as disclosed herein, in combination with another therapeutic agent.
  • the therapeutic effectiveness of a glutaminase inhibitor is enhanced by administration of another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • another therapeutic agent which also includes a therapeutic regimen
  • the overall benefit experienced by the patient is either simply additive of the two therapeutic agents or the patient experiences an enhanced (i.e., synergistic) benefit.
  • a compound disclosed herein may be appropriate to administer an agent to reduce the side effect; or the therapeutic effectiveness of a compound described herein may be enhanced by administration of an adjuvant.
  • Therapeutically effective dosages vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically effective dosages of drugs and other agents for use in combination treatment regimens are documented methodologies. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
  • the multiple therapeutic agents one of which is a glutaminase inhibitor, e.g., a GLS-1 inhibitor, as disclosed herein
  • the multiple therapeutic agents may be administered in any order, or simultaneously. If simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills).
  • one of the therapeutic agents is given in multiple doses, or both are given as multiple doses. If not simultaneous, the timing between the multiple doses optionally varies from more than zero weeks to less than twelve weeks.
  • the combination methods, compositions and formulations are not to be limited to the use of only two agents, the use of multiple therapeutic combinations are also envisioned. It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is optionally modified in accordance with a variety of factors. These factors include the disorder from which the subject suffers, as well as the age, weight, sex, diet, and medical condition of the subject. Thus, the dosage regimen actually employed varies widely, in some embodiments, and therefore deviates from the dosage regimens set forth herein.
  • the pharmaceutical agents which make up the combination therapy disclosed herein are optionally a combined dosage form or in separate dosage forms intended for substantially simultaneous administration.
  • the pharmaceutical agents that make up the combination therapy are optionally also administered sequentially, with either agent being administered by a regimen calling for two-step administration.
  • the two-step administration regimen optionally calls for sequential administration of the active agents or spaced-apart administration of the separate active agents.
  • the time between the multiple administration steps ranges from a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility,
  • a glutaminase inhibitor e.g., a GLS-1 inhibitor
  • a glutaminase inhibitor and any additional therapies are optionally administered before, during, or after the occurrence of a disease or condition, and the timing of administering the composition containing a glutaminase inhibitor, e.g., a GLS-1 inhibitor, varies in some embodiments.
  • a glutaminase inhibitor is used as a prophylactic and is administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition.
  • a glutaminase inhibitor, e.g., a GLS-1 inhibitor, and compositions are optionally
  • a glutaminase inhibitor e.g., a GLS-1 inhibitor, disclosed herein can be used in combination with anti-cancer drugs, including but not limited to the following classes: alkylating agents, anti-metabolites, plant alkaloids and terpenoids, topoisomerase inhibitors, cytotoxic antibiotics, angiogenesis inhibitors and tyrosine kinase inhibitors.
  • anti-cancer drugs including but not limited to the following classes: alkylating agents, anti-metabolites, plant alkaloids and terpenoids, topoisomerase inhibitors, cytotoxic antibiotics, angiogenesis inhibitors and tyrosine kinase inhibitors.
  • a glutaminase inhibitor may be optimally used together with one or more of the following non- limiting examples of anti-cancer agents, including, but not limited to: (1) alkylating agents, including but not limited to platinum-based agents such as cisplatin (PLATIN), carboplatin (PARAPLATIN), and oxaliplatin (ELOXATIN), as well as other alkylating agents such as streptozocin (ZANOSAR), busulfan (MYLERAN) and cyclophosphamide (ENDOXAN); (2) anti-metabolites, including but not limited to mercaptopurine
  • topoisomerase inhibitors including but not limited to irinotecan (CAMPTOSAR), topotecan (HYCAMTIN) and etoposide (EPOSIN); (5) cytotoxic antibiotics, including but not limited to actinomycin D (COSMEGEN), doxorubicin (ADRIAMYCIN), bleomycin (BLENOXANE) and mitomycin (MITOSOL); (6) angiogenesis inhibitors, including but not limited to sunitinib (SUTENT) and bevacizumab (AVASTIN); (7) tyrosine kinase inhibitors, including but not limited to imatinib (GLEEVEC), erlotinib (TARCEVA), lapatininb (TYKERB) and axitinib (INLYTA) ; 8) Hsp90 inhibitors, including but not limited to Ganetesipib; 9) MEK inhibitors, including but not limited to Trametinib and Salumetinib; 10) CD
  • mTor inhibitors including but not limited to Sirolimus and Everolimus
  • mTORCl/2 inhibitors including but not limited to AZD-8055
  • Immune Checkpoint Regulators including but not limited to Ipilimumab and Nivolumab.
  • a glutaminase inhibitor e.g., a GLS-1 inhibitor
  • a GLS-1 inhibitor e.g., a GLS-1 inhibitor
  • Therapeutic agents/treatments for treating an autoimmune and/or inflammatory condition include, but are not limited to any of the following examples: (1) corticosteroids, including but not limited to cortisone, dexamethasone, and methylprednisolone; (2) nonsteroidal anti-inflammatory drugs (NSAIDs), including but not limited to ibuprofen, naproxen, acetaminophen, aspirin, fenoprofen (NALFON), flurbiprofen (ANSAID), ketoprofen, oxaprozin (DAYPRO), diclofenac sodium
  • corticosteroids including but not limited to cortisone, dexamethasone, and methylprednisolone
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • ibuprofen including but not limited to ibuprofen, naproxen, acetaminophen, aspirin, fenoprofen (NALFON), flurbiprofen (ANSAID), keto
  • VOLTAREN diclofenac potassium
  • LODINE etodolac
  • INDOCIN indomethacin
  • TORADOL ketorolac
  • sulindac CLINORIL
  • tolmetin TOLECTIN
  • meclofenamate MECLOMEN
  • mefenamic acid PONSTEL
  • nabumetone RELAFEN
  • FELDENE piroxicam
  • immunosuppressants including but not limited to methotrexate (RHEUMATREX), leflunomide (ARAVA), azathioprine (IMURAN), cyclosporine (NEORAL, SANDIMMUNE), tacrolimus and cyclophosphamide
  • CYTOXAN (4) CD20 blockers, including but not limited to rituximab (RITUXAN); (5) Tumor Necrosis Factor (TNF) blockers, including but not limited to etanercept (ENBREL), infliximab (REMICADE) and adalimumab (HUMIRA); (6) interleukin-1 receptor antagonists, including but not limited to anakinra (KINERET); (7) interleukin-6 inhibitors, including but not limited to tocilizumab (ACTEMRA); (8) interleukin-17 inhibitors, including but not limited to AIN457; (9) Janus kinase inhibitors, including but not limited to tasocitinib; and (10) syk inhibitors, including but not limited to
  • Compounds useful in the methods of the present invention can be prepared using methods that are known to one of skill in the art. Starting materials used to prepare compounds of the present invention are commercially available or can be prepared using routine methods known in the art.
  • Compounds disclosed herein are active as GLS-1 inhibitors. Compounds disclosed in Table 1 were synthesized and tested, and had IC50s of less than ⁇ . [0233] Compounds disclosed herein are also active in inhibiting A549 cancer cell proliferation. Compounds disclosed in Table 1 were synthesized and tested, and had IC50s of less than lOOnM.
  • NRF2/KEAP1 pathway in the cancer cells is deregulated, NRF2 signaling in the cancer cells is hyperactive, the nucleic acid of the cancer cells is comprises a loss-of- function mutation in KEAP1 , the nucleic acid of said subject comprises a gain-of-function mutation in NRF2, the cancer cells have increased intracellular concentration of glutathione.
  • Cell culture media All cells were grown in RPMI-1640 (Gibcol 1875-119) with 2mM Glutamine + 10% FBS (Gibcol 6000-044) unless otherwise noted.
  • the inhibition of purified recombinant human GAC by varying concentrations of inhibitors may be assessed via a dual-coupled enzymatic assay.
  • the glutamate produced by the glutaminase reaction is used by glutamate oxidase to produce a-ketoglutarate, ammonia, and hydrogen peroxide. This hydrogen peroxide is subsequently used by horseradish peroxidase to produce resorufin in the presence of Amplex UltraRed.
  • the assay buffer consisted of 50mM Hepes (pH 7.4), 0.25mM EDTA and O. lmM Triton X-100.
  • GAC was incubated with potassium phosphate (10 minutes at room temperature) prior to incubation with inhibitor (10 minutes at room temperature).
  • the final reaction conditions were as follows: 2nM GAC, 50mM potassium phosphate, lOOmU/mL glutamate oxidase (Sigma), lmM glutamine (Sigma), lOOmU/mL
  • A549 cells were routinely maintained in RPMI 1640 media (Gibco catalog number 1 1875-093) supplemented with 10% dialyzed fetal bovine serum using a humidified incubator (37oC, 5% CO2 and ambient O2).
  • RPMI 1640 media Gibco catalog number 1 1875-093
  • 10% dialyzed fetal bovine serum using a humidified incubator (37oC, 5% CO2 and ambient O2).
  • cells were inoculated into 384-well black CulturPlates (Perkin Elmer) at a density of 1000 cells/well in a volume of 40uL.
  • 384-well black CulturPlates Perkin Elmer
  • cells were treated with compound (lOuL) in a final DMSO concentration of 0.5% (v/v).
  • the microplates were then incubated for 72 hours (37 °C, 5% CO2 and ambient O2).
  • A549 cells were plated at 400,000 cells per well in a 6-well dish. H727 cells were plated at 800,000 cells per well in a 6 well dish. 24h after plating, media was removed and replaced with media containing ⁇ ⁇ IACS-01 1393 or 0.01%DMSO (control). A Oh time point was immediately collected by removing 250 ⁇ 1 of media from each well into a 1.7mL Eppendorf tube. These samples were placed at -20 for storage. Subsequent collections for each time point were made in the same fashion. All samples are stored at -20 until the analysis step. At 24h, an additional 250 ⁇ 1 of media was collected from each sample. Media was frozen after collection.
  • cell lysates were collected through lysis with glycerol lysis buffer (see below) by adding 200ul of lysis buffer to each well, incubating at 37 degrees for 5 min, and collecting all lysate into a microfuge tube. At each time point, one well for each condition was fixed and stained with crystal violet to normalize for cell number (see protocol below) .All crystal violet samples were solubilized after the final timepoint was collected and used to normalize the collected data. Collected samples were analyzed for glutamine and glutamate content using the YSI2950 Bio-analyzer equipped with membranes from measure the relevant chemistries.
  • the plate was left upside down, uncovered for at least 5 hours to dry. Once completely dry, it was solubilized in 10% acetic acid by adding ⁇ to all stained wells. After 5 minutes, all wells were pipet mixed and after another 5 minutes read on Pherastar at OD 590nm.
  • A549 cells were plated at 4.5xl0 6 cells per 10cm dish 24h prior to treatment. Cells were treated on day zero (d.0) at time zero (t.0) with 0.01% DMSO, ⁇ , ⁇ , or ⁇ of IACS-011393 for 24h. On day 1 (d. l), 2 hours prior to harvest, samples were washed with 5mL media containing 10% dialyzed FBS.lOmL of media was added to plates and plates were returned to the incubator for 2h. Media was then aspirated from plates and 4mL of 80% chilled MeOH (-80°C) was immediately added. Plates were incubated at -80°C for 15 minutes.
  • A549 cells were plated at 4.5xl0 5 cells per 6cm dish 24h prior to treatment. On the day of the experiment, cells were treated with 0.01% DMSO, or ⁇ of IACS- 011393 for 6h in the presence or absence of fully labeled 13 C-Glutamine (Cambridge Isotope Laboratories, Inc., CLM01822). Labeled glutamine was added in regular media containing no unlabeled glutamine, such that cells could only metabolize the labeled form. After treatment, media was then aspirated from plates, plates were washed 2x in cold PBS, and 500 ⁇ of 80% chilled MeOH (-80°C) was immediately added. Plates were incubated at -80°C for 15 minutes.
  • Samples (10 ⁇ ) were injected into a Waters XBridge Amide column for HILIC/MS analysis.
  • the mobile phases A, 20 mM ammonium acetate, 20 mM acetic acid in 95% water/acetonitrile and B, 100% acetonitrile were used with a linear gradient elution from 85% B to 100 % A for a total sample run time of 30 min.
  • the raw data was processed using Agilent Mass Hunter Qualitative Analysis. Isotopologues of putative metabolites incorporating carbons derived from [U- 13 C]-glutamine were identified based on their accurate masses and retention times which were further confirmed with those of authentic standards and public database compound libraries.
  • ssGSEA Single sample gene set enrichment
  • CCLE Cancer Cell Line Encyclopedia
  • the NRF2 signature gene set was based on a known set of NRF2 target genes: ABCC1, ABCC2, G6PD, GCLC, GCLM, GR 6, GSR, GSTM4, HMOX1, ME1, MGST1, NQOl, PRDX1, TXN and TXNRD1.
  • Affymetrix U133 Plus2.0 microarrays were performed for each condition in triplicates.
  • Robust multi-array average (RMA) method was used with default options (with background correction, quantile normalization, and log transformation) to normalize raw data from batches using R/Bioconductor's affy package (Irizarry et al 2003).
  • RMA Robust multi-array average
  • Differentially expression analysis was carried out using a moderated t-test function from Bioconductor's limma package (Smyth 2005). A gene is called as differentially expressed if FDR corrected p-value is less than 0.05.
  • A549 cells were seeded in 6 well dishes (250,000 cells per well). The day of the experiment, cells were treated with 0.01% DMSO or ⁇ IACS-011393 for 24 hours. The following day, an allocated well was treated with hydrogen peroxide for 20-30 minutes as a positive control for ROS generation. All wells were then stained with CM-H2DCFDA (5 ⁇ , Life Technologies) for 30 minutes at 37°C, washed with IX PBS, harvested by trypsinization, and re- suspended in 400 ⁇ phenol-free RPMI medium. Cells were strained through a 40 ⁇ cell strainer and analyzed using a flow cytometer (LSR Fortessa). Values displayed are fold changes compared to DMSO for mean fluorescence intensity of entire population.
  • Example 1 NSCLC cell lines are differentially sensitive to GLS inhibition.
  • the chemical probe inhibits the conversion of glutamine to glutamate and reduces glutamine-dependent oxygen consumption, both measures of GLS-dependent activity (Fig. IB, 1C).
  • Figure IB shows that target engagement can be measured after treatment with IACS-011393.
  • the conversion of glutamine to glutamate was measured in extracellular media and cell lysates in samples treated with DMSO (control) or IACS-011393.
  • IACS-011393 Treatment of cells for 24 hours with IACS-011393 prevented the conversion of glutamine to glutamate (as measured by the ratio of glutamate to glutamine (GLU:GLN)). This inhibition was evident by a decrease in the GLU:GLN ratio outside the cell and in cell lysates.
  • FIG. 1C shows that target A549 cells utilize glutamine anaplerosis to drive respiration.
  • A549 cells were plated and then starved of nutrients for 60 minutes. After starvation, cells were pre-loaded with the indicated concentrations of IACS-011393 to inhibit GLS. Next, 2mM glutamine was added to the wells to trigger oxygen
  • IACS-011393 shows a dose-dependent inhibition of glutamine - dependent oxygen consumption, suggesting that A549 cells utilize glutamine anaplerosis as a metabolite source for the TCA cycle. This process is blocked by the glutaminase inhibitor ("GLSi").
  • FIG. ID shows differential sensitivity to IACS-011393 is seen in various NSCLC lines.
  • A549, NCI-H2122, H1650 and H1395 NSCLC lines were treated with IACS-011393 as in (A) above and 72 hour viability was analyzed. While A549 and H2122 show nM sensitivity to GLSi, H1395 and H1650 cells are resistant to treatment with GLSi.
  • Example 2 GLS inhibition alters redox balance in responder cell lines.
  • AICAR aminoimidazole carboxamide ribonucleotide
  • IMP inosine monophosphate
  • R5P ribose 5-phosphate
  • S7P sedoheptulose-l-7-phosphate
  • SBP sedoheptulose-l,7-bisphosphate
  • G3P glyceraldehdye-3 -phosphate
  • DHAP dihydroxy-acetone-phosphate
  • E4P erythrose-4- phosphate
  • A549 cells were grown in the presence of DMSO or IACS-011393 in media containing fully labeled 13 C-glutamine as the sole glutamine source. After 6 hours, cell lysates were extracted and analyzed by LC/MS to examine the incorporation of stably labeled carbons into metabolites within cells. As shown in the left panel, fully labeled glutamate is present in cells that have been treated with DMSO. However, in cells treated with IACS- 011393, there are no labeled carbons in the glutamate pools remaining in the cell (m+5, white bars).
  • DMSO treated cells contain labeled carbons in their pools of glutathione, while cells treated with IACS-011393 do not.
  • FIG. 2C shows a schematic representation of the incorporation of carbons from fully labelled 13C-Glutamine into glutathione in the absence or presence of GLSi -
  • Fig. 2B A schematic representation of the results presented in Fig. 2B. Dark circles represent labelled carbon atoms in cells with functional glutaminase (left) or cells treated with a glutaminase inhibitor (right).
  • NRF2/KEAP1 pathway modulation confers sensitivity to GLS inhibition.
  • NRF2 is a transcription factor that binds the Antioxidant Response Element (ARE) and initiates transcription of a number of genes involved in regulation of redox balance and cellular detoxification.
  • ARE Antioxidant Response Element
  • KEAPl is a negative regulator of NRF2, maintaining activity through induced proteosomal degradation.
  • Nrf2 score is a measure of the relative expression of Nrf2 target genes.
  • this pathway is hyperactivated through loss of function mutations in KEAPl or gain of function mutations in NRF2 which limit or prevent the interaction of these two proteins (Fig. 3B).
  • the mutations in responder cell lines fall within important functional domains of Keapl and Nrf2 as shown in a schematic representation of example mutations in Keapl and Nrf2 in cell lines tested in Fig. 3 A. Most mutations fall within important functional domains that regulate Keapl activity, or Keapl -Nrf2 binding.
  • the exception in the cell lines tested is the D77N mutation in NCI-H1568 cells which falls outside of one of two Keapl -binding domains in Nrf2.
  • Target engagement is seen in both responder and non-responder cell lines (Fig. 3C).
  • Treatment of cells with IACS-011393 results in decreased conversion of glutamine to glutamate as monitored by the intracellular ratio of GLU:GLN in both responder and non-responder cell lines.
  • Representative data is shown in Figure 3C for A549 cells (a responder line) and H727 cells (a non-responder line) suggesting that responder lines are hyper-dependent on GLS-mediated glutaminolysis, while non- responder lines are less dependent on this process.
  • NRF2 NRF2 drives the expression of Glutamate-cysteine ligase (GCLC), the rate limiting enzyme that catalyzes glutathione synthesis from intracellular pools of glutamate and cysteine.
  • GCLC Glutamate-cysteine ligase
  • NRF2/KEAP1 mutant cells have evolved a dependence on glutathione to regulate redox balance.
  • GLS and glutathione synthesis are inhibited, cells are unable to produce sufficient free radical scavengers to maintain redox balance within the cell.
  • Figure 4B shows that the loss of glutathione after treatment with a
  • ROS reactive oxygen species
  • FIG. 4C shows accumulation of DNA damage after IACS-011393 treatment can be rescued by application of cell-permeable glutathione.
  • A549 cells treated with either DMSO or IACS-011393 for 48 hours were stained with Hoechst's Stain (nuclei) and an antibody to ⁇ 2 ⁇ (DNA damage puncta) and then analyzed by high-content imaging.
  • ⁇ 2 ⁇ foci were counted and quantified per nuclei. Quantification (representative images shown) reveals that IACS-011393 treatment induces DNA damage in A549 cells.
  • Exogenous application of 4mM cell-permeable glutathione reduces the accumulation of DNA damage, presumably by scavenging ROS that has accumulated after IACS-011393 treatment.
  • FIG. 4D shows the results of A549 and H2122 cells that were treated with IACS-011393 at the indicated doses in a 72h growth assay.
  • a subset of the IACS-011393 treated cells received applications of cell-permeable GSH (once at the time of IACS-011393 treatment, once 24h post-treatment and once 48h post-treatment).
  • Application of exogenous GSH rescued a significant portion of cell growth that was inhibited after IACS-011393 treatment.
  • This data supports the idea that Keapl/Nrf2 mutant cell lines are especially sensitive to GLSi due to a heavy dependence on Nrf2-driven glutathione pools that allow these cells to grow under conditions of extreme oxidative stress.
  • NRF2/KEAP1 mutant NSCLC we challenged cells with an IC90 dose (lOOnM) of IACS- 11393 for 8 weeks until resistant clones emerged. Rechallenge of either the bulk population (Fig. 5A) or selected single clones (Fig. 5B) confirmed a >3 log shift in IC50 values.
  • Fig. 5C comprehensive metabolomic
  • Fig. 5D transriptomic analyses
  • Fig. 5 A shows that cells treated chronically with GLSi acquire resistance to IACS-011393. Bulk populations of A549 cells were carried in media containing increasing concentrations of GLSi over the course of two months up to a high
  • Fig. 5B shows that clones resistant to GLSi display activation of alternative pathways to maintain redox balance. Clones were subjected to baseline metabolomics analysis and compared to the parental A549 cell line. Log2-fold changes are shown for metabolites that were significantly up or down-regulated in comparison to baseline levels in the parental strain. These data indicate that (a) that clones have developed alternative sources of glutamate; (b) cells have activated alternative pathways to generate reducing power; (c) pathways that may contribute intermediates to the TCA cycle independent of glutaminolysis have been up regulated. [0273] Fig. 5C shows that clones resistant to GLSi display altered transcriptional profiles to produce alternative sources of glutamate and reducing power.
  • Fig. 5D provides a global overview of acquired adaptation to glutaminase inhibition.
  • a global overview of metabolic and transcriptomic changes observed in cells resistant to chronic GLSi shows that A549 cells are able to initiate multiple pathways that provide reducing power to cells that are unable to convert glutamine to glutamate and glutathione through the activity of glutaminase.
  • the main themes are alternative sources of glutamate, which may be used for glutathione synthesis and anaplerosis, along with an up-regulation of pathways involved in generating reducing intermediate (e.g., NADPH from malic enzyme).
  • Bolded text represents metabolites or transcripts that were up relative to parental cells, while italicized text represents metabolites or transcripts that were down relative to parental cells.
  • SAM S-adenosyl-L-methionine
  • SAH S- adenosyl-L-homocysteine
  • hcys homocysteine
  • AICAR aminoimidazole carboxamide ribonucleotide
  • IMP inosine monophosphate
  • R5P ribose 5-phosphate
  • S7P sedoheptulose-l-7
  • Example 6 NRF2/KEAP1 deregulation sensitizes cancer cells to GLS inhibition.
  • the NRF2/KEAP1 pathway regulates redox balance in part through the transcriptional regulation of GCLC, the enzyme required to convert glutamine-derived glutamate and cysteine into glutathione (Fig. 6).
  • Figure 6 shows that cells with loss-of- function mutations in KEAPlor gain-of- function mutations in NRF2 become addicted to high levels of glutathione for coping with oxidative stress during conditions of rapid proliferation (Tumor Cell, Fig. 6A).
  • Tumor Cell Fig. 6B
  • intracellular pools of glutamate are drastically reduced, and thus, glutathione synthesis is inhibited. This response shifts the redox balance of tumor cells causing an increase in ROS accumulation and DNA damage leading to cell death.
  • glutathione provides reducing power to limit the mutagenic and DNA damaging effects of intracellular ROS.
  • Tumors that harbor somatic mutations that deregulate the NRF2/KEAP1 pathway evolve a dependence on glutathione and an addiction on glutamine.
  • GLS inhibition reduces the conversion of glutamine to glutamate and thus inhibits the synthesis of glutathione.
  • NRF2/KEAP1 mutant cells accumulate ROS which induces DNA damage and induces cell death. This model explains the extraordinarily of NRF2/KEAP1 mutant NSCLC cell lines to GLS inhibition.

Abstract

L'invention concerne des procédés de traitement d'un patient qui présente une modification de la voie NRF2/KEAP1, et des composés et des compositions utiles pour un tel traitement. L'invention concerne également des procédés permettant d'évaluer s'il convient d'administrer à un patient un composé qui inhibe la production de glutathion ou un inhibiteur de glutaminase.
PCT/US2015/039153 2014-07-03 2015-07-03 Thérapie par inhibiteur de glutaminase WO2016004418A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
ES15814655T ES2921989T3 (es) 2014-07-03 2015-07-03 Terapia con inhibidores de glutaminasa
EP15814655.5A EP3164195B1 (fr) 2014-07-03 2015-07-03 Thérapie par inhibiteur de glutaminase
DK15814655.5T DK3164195T3 (da) 2014-07-03 2015-07-03 Glutaminaseinhibitorterapi

Applications Claiming Priority (16)

Application Number Priority Date Filing Date Title
US201462020519P 2014-07-03 2014-07-03
US201462020539P 2014-07-03 2014-07-03
US201462020731P 2014-07-03 2014-07-03
US201462020524P 2014-07-03 2014-07-03
US62/020,524 2014-07-03
US62/020,731 2014-07-03
US62/020,519 2014-07-03
US62/020,539 2014-07-03
US201562187160P 2015-06-30 2015-06-30
US62/187,160 2015-06-30
US14/791,206 2015-07-02
US14/791,186 2015-07-02
US14/791,206 US20160002248A1 (en) 2014-07-03 2015-07-02 Gls1 inhibitors for treating disease
US14/791,186 US9809588B2 (en) 2014-07-03 2015-07-02 GLS1 inhibitors for treating disease
US14/791,284 US20160002204A1 (en) 2014-07-03 2015-07-03 Gls1 inhibitors for treating disease
US14/791,284 2015-07-03

Publications (1)

Publication Number Publication Date
WO2016004418A1 true WO2016004418A1 (fr) 2016-01-07

Family

ID=55020035

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/039153 WO2016004418A1 (fr) 2014-07-03 2015-07-03 Thérapie par inhibiteur de glutaminase

Country Status (1)

Country Link
WO (1) WO2016004418A1 (fr)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016160980A1 (fr) * 2015-03-30 2016-10-06 Calithera Biosciences, Inc. Procédés d'administration d'inhibiteurs de glutaminase
US9687485B2 (en) 2014-06-13 2017-06-27 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
US9809588B2 (en) 2014-07-03 2017-11-07 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
WO2018039544A1 (fr) * 2016-08-25 2018-03-01 Calithera Biosciences, Inc. Polythérapie comprenant des inhibiteurs de glutaminase
US9938267B2 (en) 2011-11-21 2018-04-10 Calithera Biosciences, Inc. Heterocyclic inhibitors of glutaminase
CN108676727A (zh) * 2018-05-14 2018-10-19 浙江海洋大学 一种具有Keap1-Nrf2通路抑制活性的海洋真菌
US10125128B2 (en) 2015-06-30 2018-11-13 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US10150753B2 (en) 2015-12-22 2018-12-11 Board Of Regents, The University Of Texas System Salt forms and polymorphs of (R)-1-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)-6-methylpyrdin-2-yl)acetamido) pyridazin-3-yl)-2-fluorobutyl)-N-methyl-1H-1,2,3-triazole-4-carboxamide
US10195197B2 (en) 2016-08-25 2019-02-05 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
US10258619B2 (en) 2015-10-05 2019-04-16 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors and immuno-oncology agents
WO2019079632A1 (fr) 2017-10-18 2019-04-25 Board Of Regents, The University Of Texas System Thérapie par inhibiteur de glutaminase
US10316030B2 (en) 2014-08-07 2019-06-11 Calithera Biosciences, Inc. Crystal forms of glutaminase inhibitors
US10441587B2 (en) 2015-04-06 2019-10-15 Calithera Biosciences, Inc. Treatment of lung cancer with inhibitors of glutaminase
CN110746416A (zh) * 2019-09-05 2020-02-04 中国药科大学 含有三氮唑结构的谷氨酰胺酶gls1抑制剂或其可药用的盐、其制备方法及用途
WO2022122044A1 (fr) * 2020-12-11 2022-06-16 杭州紫晶医药科技有限公司 Composé hétérocyclique servant d'inhibiteur de gls1

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100255117A1 (en) * 2007-04-06 2010-10-07 The Johns Hopkins University Methods and compositions for the treatment of cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100255117A1 (en) * 2007-04-06 2010-10-07 The Johns Hopkins University Methods and compositions for the treatment of cancer

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
HOOVER, JOHN E.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
INAMI ET AL.: "Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells", THE JOURNAL OF CELL BIOLOGY, vol. 193, no. 2, 1 April 2011 (2011-04-01), pages 275 - 284, XP055158643 *
KIBBE ET AL.: "Handbook of Pharmaceutical Excipients", 1999, AMERICAN PHARMACEUTICAL ASSOCIATION
LI ET AL.: "Sulforaphane Potentiates the Efficacy of 17-Allylamino 17- Demethoxygeldanamycin Against Pancreatic Cancer Through Enhanced Abrogation of Hsp90 Chaperone Function", NUTRITION AND CANCER, vol. 63, no. 7, 29 August 2011 (2011-08-29), pages 1151 - 1159, XP055167208 *
LIBERMAN ET AL.: "Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
ROBINSON ET AL.: "Novel mechanism of inhibition of rat kidney-type glutaminase by bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES).", THE BIOCHEMICAL JOURNAL, vol. 406, 15 September 2007 (2007-09-15), pages 407 - 414, XP055069615 *
ROTBLAT ET AL.: "NRF2 and p53: Januses in cancer?", ONCOTARGET, vol. 3, no. 11, 19 November 2012 (2012-11-19), pages 1272 - 1283, XP055251330 *
See also references of EP3164195A4 *
SINGH ET AL.: "Dysfunctional KEAP1-NRF2 interactions in non-small cell lung cancer", PLOS MEDICINE, vol. 3, no. 10, October 2006 (2006-10-01), pages 1865 - 1876, XP003015746, DOI: 10.1371/journal.pmed.0030420
STAHL, P. HEINRICH: "Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCHA
THANGAVELU ET AL.: "Structural basis for the allosteric inhibitory mechanism of human kidney-type glutaminase (KGA) and its regulation by Raf-Mek-Erk signaling in cancer cell metabolism", PROC. NATL. ACAD. SCI. USA, vol. 109, no. 20, 15 May 2012 (2012-05-15), pages 7705 - 7710, XP055251346 *
ZHANG ET AL.: "Distinct Cysteine Residues in Keap1 Are Required for Keap1-Dependent Ubiquitination of Nrf2 and for Stabilization of Nrf2 by Chemopreventive Agents and Oxidative Stress", MOLECULAR AND CELLULAR BIOLOGY, vol. 23, no. 22, 1 November 2003 (2003-11-01), pages 8137 - 8151, XP055251345 *
ZHANG ET AL.: "Loss of Kelch-Like ECH-Associated Protein 1 Function in Prostate Cancer Cells Causes Chemoresistance and Radioresistance and Promotes Tumor Growth", MOLECULAR CANCER THERAPEUTICS, vol. 9, no. 2, 1 February 2010 (2010-02-01), pages 336 - 347, XP055227547 *

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9938267B2 (en) 2011-11-21 2018-04-10 Calithera Biosciences, Inc. Heterocyclic inhibitors of glutaminase
US9687485B2 (en) 2014-06-13 2017-06-27 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
US11370786B2 (en) 2014-07-03 2022-06-28 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US9809588B2 (en) 2014-07-03 2017-11-07 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US11958849B2 (en) 2014-07-03 2024-04-16 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US10766892B2 (en) 2014-07-03 2020-09-08 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US10344025B2 (en) 2014-07-03 2019-07-09 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US10676472B2 (en) 2014-08-07 2020-06-09 Calithera Biosciences, Inc. Crystal forms of glutaminase inhibitors
US10316030B2 (en) 2014-08-07 2019-06-11 Calithera Biosciences, Inc. Crystal forms of glutaminase inhibitors
CN107921031A (zh) * 2015-03-30 2018-04-17 卡利泰拉生物科技公司 给予谷氨酰胺酶抑制剂的方法
EA037152B1 (ru) * 2015-03-30 2021-02-11 Калитера Байосайенсиз, Инк. Способ лечения рака
WO2016160980A1 (fr) * 2015-03-30 2016-10-06 Calithera Biosciences, Inc. Procédés d'administration d'inhibiteurs de glutaminase
US10441587B2 (en) 2015-04-06 2019-10-15 Calithera Biosciences, Inc. Treatment of lung cancer with inhibitors of glutaminase
US10125128B2 (en) 2015-06-30 2018-11-13 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US11713313B2 (en) 2015-06-30 2023-08-01 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
AU2016287661B2 (en) * 2015-06-30 2020-07-16 Board Of Regents, University Of Texas System GLS1 inhibitors for treating disease
US10738043B2 (en) 2015-06-30 2020-08-11 Board Of Regents, The University Of Texas System GLS1 inhibitors for treating disease
US10258619B2 (en) 2015-10-05 2019-04-16 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors and immuno-oncology agents
US10940148B2 (en) 2015-10-05 2021-03-09 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors and immuno-oncology agents
US11603365B2 (en) 2015-12-22 2023-03-14 Board Of Regents, The University Of Texas System Salt forms and polymorphs of (r)-1-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)-6-methylpyridin-2-yl)acetamido) pyridazin-3-yl)-2-fluorobutyl)-n-methyl-1H-1,2,3-triazole-4-carboxamide
US10150753B2 (en) 2015-12-22 2018-12-11 Board Of Regents, The University Of Texas System Salt forms and polymorphs of (R)-1-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)-6-methylpyrdin-2-yl)acetamido) pyridazin-3-yl)-2-fluorobutyl)-N-methyl-1H-1,2,3-triazole-4-carboxamide
US10899740B2 (en) 2015-12-22 2021-01-26 Board Of Regents, The University Of Texas System Salt forms and polymorphs of (R)-1-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)-6-methylpyridin-2-yl)acetamido)pyridazin-3-yl)-2-fluorobutyl)-N-methyl-1H-1,2,3-triazole-4-carboxamide
US10195197B2 (en) 2016-08-25 2019-02-05 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
US10278968B2 (en) 2016-08-25 2019-05-07 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
WO2018039544A1 (fr) * 2016-08-25 2018-03-01 Calithera Biosciences, Inc. Polythérapie comprenant des inhibiteurs de glutaminase
WO2019079632A1 (fr) 2017-10-18 2019-04-25 Board Of Regents, The University Of Texas System Thérapie par inhibiteur de glutaminase
US11045443B2 (en) 2017-10-18 2021-06-29 Board Of Regents, The University Of Texas System Glutaminase inhibitor therapy
EP3697764A4 (fr) * 2017-10-18 2021-07-21 Board Of Regents, The University Of Texas System Thérapie par inhibiteur de glutaminase
CN111225903A (zh) * 2017-10-18 2020-06-02 德州大学系统董事会 谷氨酰胺酶抑制剂疗法
JP2021500335A (ja) * 2017-10-18 2021-01-07 ボード オブ レジェンツ, ザ ユニバーシティ オブ テキサス システムBoard Of Regents, The University Of Texas System グルタミナーゼ阻害薬療法
JP7361687B2 (ja) 2017-10-18 2023-10-16 ボード オブ レジェンツ,ザ ユニバーシティ オブ テキサス システム グルタミナーゼ阻害薬療法
US11786500B2 (en) 2017-10-18 2023-10-17 Board Of Regents, The University Of Texas System Glutaminase inhibitor therapy
US10722487B2 (en) 2017-10-18 2020-07-28 Board Of Regents, The University Of Texas System Glutaminase inhibitor therapy
CN108676727A (zh) * 2018-05-14 2018-10-19 浙江海洋大学 一种具有Keap1-Nrf2通路抑制活性的海洋真菌
CN110746416A (zh) * 2019-09-05 2020-02-04 中国药科大学 含有三氮唑结构的谷氨酰胺酶gls1抑制剂或其可药用的盐、其制备方法及用途
WO2021042723A1 (fr) * 2019-09-05 2021-03-11 中国药科大学 Inhibiteur de glutaminase gls1 contenant une structure triazole ou un sel pharmaceutiquement acceptable de celui-ci, procédé de préparation correspondant et utilisation associée
WO2022122044A1 (fr) * 2020-12-11 2022-06-16 杭州紫晶医药科技有限公司 Composé hétérocyclique servant d'inhibiteur de gls1

Similar Documents

Publication Publication Date Title
WO2016004418A1 (fr) Thérapie par inhibiteur de glutaminase
US20190134032A1 (en) Glutaminase inhibitor therapy
US11786500B2 (en) Glutaminase inhibitor therapy
EP3833670B1 (fr) Dérivés de 6-(4-amino-3-méthyl-2-oxa-8-azaspiro[4.5]décan-8-yl)-3-(2,3-dichlorophényl)-2-méthylpyrimidin-4(3h)-one et composés similaires et tant qu'inhibiteurs ptpn11 (shp2) pour le traitement du cancer
US11840536B2 (en) Heterocyclic inhibitors of PTPN11
US11058688B2 (en) Imidazopiperazine inhibitors of transcription activating proteins
AU2016287661B2 (en) GLS1 inhibitors for treating disease
CA3137901A1 (fr) Inhibiteurs heterocycliques de tyrosine kinase
WO2015075165A1 (fr) Inhibiteurs de translation en chimiothérapie à dose élevée et/ou radiothérapie à dose élevée
WO2020219906A1 (fr) Inhibiteurs hétérocycliques de tyrosine kinase
EP3164195A1 (fr) Thérapie par inhibiteur de glutaminase
US20230312588A1 (en) Imidazopiperazine inhibitors of transcription activating proteins
US20230295173A1 (en) Imidazopiperazine inhibitors of transcription activating proteins
US11466017B2 (en) Heterocyclic inhibitors of PTPN11
US20220257601A1 (en) Inhibitors of prc1 for treatment of cancer

Legal Events

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

Ref document number: 15814655

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2015814655

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015814655

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE