US20180221395A1 - Methods for cancer and immunotherapy using prodrugs of glutamine analogs - Google Patents

Methods for cancer and immunotherapy using prodrugs of glutamine analogs Download PDF

Info

Publication number
US20180221395A1
US20180221395A1 US15/885,275 US201815885275A US2018221395A1 US 20180221395 A1 US20180221395 A1 US 20180221395A1 US 201815885275 A US201815885275 A US 201815885275A US 2018221395 A1 US2018221395 A1 US 2018221395A1
Authority
US
United States
Prior art keywords
cancer
alkyl
group
substituted
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/885,275
Other languages
English (en)
Inventor
Barbara Slusher
Jonathan Powell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Johns Hopkins University
Original Assignee
Johns Hopkins University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Johns Hopkins University filed Critical Johns Hopkins University
Priority to US15/885,275 priority Critical patent/US20180221395A1/en
Publication of US20180221395A1 publication Critical patent/US20180221395A1/en
Priority to US16/262,476 priority patent/US10842763B2/en
Priority to US17/102,341 priority patent/US11759444B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/223Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of alpha-aminoacids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/225Polycarboxylic acids
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/655Azo (—N=N—), diazo (=N2), azoxy (>N—O—N< or N(=O)—N<), azido (—N3) or diazoamino (—N=N—N<) compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/664Amides of phosphorus acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • A61K31/685Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols one of the hydroxy compounds having nitrogen atoms, e.g. phosphatidylserine, lecithin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46433Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46434Antigens related to induction of tolerance to non-self
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Cells under certain conditions may undergo a metabolic switch from a metabolic profile that requires less activity of certain metabolic pathways to meet the cell's energy demands to a metabolic profile that requires greater activity of those metabolic pathways or increased activity of other metabolic pathways to meet its energy demands.
  • cells under certain conditions may undergo a switch toward increased glycolysis and away from oxidative phosphorylation (OXPHOS).
  • OXPHOS oxidative phosphorylation
  • glycolysis provides less adenosine triphosphate (ATP) than oxidative phosphorylation
  • ATP adenosine triphosphate
  • aerobic glycolysis permits the generation of the substrates necessary for the generation of amino acids, nucleic acids and lipids, all of which are crucial for proliferation (Vander Heiden et al. (2009) Science 324(5930):1029-1033).
  • RNA interference RNA interference
  • the presently disclosed subject matter provides a method for treating a cancer in a subject in need thereof, the method comprising: (a) administering a therapeutically effective amount of a first immunotherapy to the subject, wherein the first immunotherapy is a metabolic reprogramming agent that decreases glutamine metabolic activity; and (b) optionally administering a therapeutically effective amount of a second immunotherapy to the subject.
  • the metabolic reprogramming agent is a glutamine antagonist.
  • the metabolic reprogramming agent is a glutamine analog that interferes with a glutamine metabolic pathway.
  • the metabolic reprogramming agent is selected from the group consisting of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV).
  • the metabolic reprogramming agent is a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway.
  • at least one metabolic reprogramming agent is a prodrug of acivicin, azaserine, DON, and L-DONV.
  • at least one metabolic reprogramming agent is a compound having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy, e.g., an immunotherapeutic agent, to the subject.
  • a therapeutically effective amount of the second immunotherapy e.g., an immunotherapeutic agent
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an immune checkpoint blockade therapy.
  • the immune checkpoint blockade therapy is selected from the group consisting of PD-1 antagonists, PD-L1 antagonists, CTLA-4 antagonists, LAG3 antagonists, B7-H3 antagonists, and combinations thereof.
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an adoptive cellular therapy.
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is marrow-infiltrating lymphocytes (MILs).
  • MILs marrow-infiltrating lymphocytes
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an adenosine A2aR blockade.
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a tumor vaccine.
  • the method includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a passive immunotherapy antibody.
  • the passive immunotherapy antibody is selected from the group consisting of bevacizumab, cetuximab, rituximab, trastuzumab, alemtuzumab, ibritumomab tiuxetan, panitumumab, and combinations thereof.
  • the method includes simultaneously or sequentially administering to the subject a therapeutically effective amount of a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and combinations thereof.
  • a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and combinations thereof.
  • the first immunotherapy, and the second immunotherapy if administered is/are administered to the subject in the absence of a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and combinations thereof.
  • a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and combinations thereof.
  • the cancer is: (i) a cancer of the central nervous system; (ii) a cancer that is associated with transplant and/or immunosuppression; (iii) a cancer that is refractory to chemotherapy; (iv) a cancer that is refractory to photodynamic therapy; (v) a cancer that is refractory to proton therapy; (vi) a cancer that is refractory to radiotherapy; and (vii) a cancer that is refractory to surgery.
  • the cancer is a newly diagnosed, recurrent, and/or refractory cancer selected from the group consisting of celnasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T-cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger-Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter cancer, brain cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer, osteochondroma, chondrosarcoma, Ewing's sarcoma
  • the presently disclosed subject matter provides a method of preventing or reducing the incidence of a relapse in a cancer subject in remission, the method comprising administering to the subject a therapeutically effective amount of a metabolic reprogramming agent, wherein the metabolic reprogramming agent is selected from the group consisting of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV), and prodrugs thereof.
  • at least one metabolic reprogramming agent is a compound having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.
  • the metabolic reprogramming agent is: (i) administered to the subject post transplant; (ii) administered to the subject post chemotherapy; (iii) administered to the subject post immunotherapy; (iv) administered to the subject post photodynamic therapy; (v) administered to the subject post proton therapy; (vi) administered to the subject post radiotherapy; (vii) administered to the subject post surgery; and combinations thereof.
  • FIG. 1 is a line graph showing that metabolic reprogramming therapy with at least one metabolic reprogramming agent (e.g., DON) markedly inhibits lymphoma growth in a EL4 mouse lymphoma model, suggesting that bone marrow derived tumors may be extraordinarly susceptible to metabolic reprogramming therapy with at least one metabolic reprogramming agent (e.g., DON);
  • at least one metabolic reprogramming agent e.g., DON
  • FIG. 2 is a bar graph showing that metabolic reprogramming therapy with at least one metabolic reprogramming agent (e.g., DON) has a modest effect on inhibiting melanoma growth, which is a not a bone marrow derived tumor;
  • DON metabolic reprogramming agent
  • FIG. 3 is a graph showing that metabolic reprogramming therapy with at least one metabolic reprogramming agent (e.g., DON) conditions B16 melanoma to be killed by immunotherapy by inhibiting tumor infiltrating Regulatory T cells (Foxp3 + );
  • at least one metabolic reprogramming agent e.g., DON
  • FIG. 4 is an illustration showing the structures of DON and DON-based prodrugs
  • FIG. 5A is a bar graph and FIG. 5B is a line graph showing that DON (1) inhibits glutamine metabolism and GBM tumor growth in vivo.
  • FIG. 5A shows compound 1 (0.8 mg/kg, i.p.) inhibited glutamine metabolism as evidenced by increased endogenous glutamine concentrations in flank GBM tumors 2 hours post-administration relative to vehicle-treated controls; *p ⁇ 0.05.
  • FIG. 5B shows in efficacy studies, compared to Day 0 baseline, vehicle-treated mice exhibited significant growth of flank GBM tumors during the course of the experiment.
  • systemic administration of 1 (0.8 mg/kg, i.p, q.d. days 1-6) caused a dramatic reduction in tumor size; ***p ⁇ 0.001, ****p ⁇ 0.0001.
  • Table 1 Note the bold numbers following the terms “DON”, “DON prodrugs”, “DON-based prodrugs”, and the like, refer to particular compounds disclosed in Table 1 below.
  • FIGS. 6A and 6B are lines graphs and FIG. 6C is a table showing in vivo brain and plasma pharmacokinetics of compound DON (1) following oral administration of DON (1) and 5c in mice.
  • 1 and 5c were dosed in mice at 0.8 mg/kg equivalent, via oral gavage and plasma and brain concentrations of compound 1 were evaluated via LC/MS.
  • Oral administration of compound 1 and 5c exhibited similar plasma and brain pharmacokinetic profiles due to complete and rapid metabolism of 5c to 1 in mouse plasma;
  • FIG. 7A is a line graph
  • FIG. 7B is a bar graph
  • FIG. 7C is a table
  • an FIG. 7D is an illustration showing in vivo pharmacokinetics of DON following i.v. administration of DON (1) and 5c in monkey plasma and CSF.
  • 1 and 5c were dosed in two pigtail macaques at 1.6 mg/kg equivalent of 1 via i.v. administration and plasma (0.25-6 h) and CSF (30 min) concentrations of DON were evaluated via LC/MS.
  • Relative to 1 5c delivered substantially lower DON plasma concentration.
  • FIG. 8A is a Kaplan-Meier graph
  • FIG. 8B , FIG. 8C , FIG. 8D , and FIG. 8E are line graphs showing that 25 (5 day dosing starting on day 7) is superior to CB-839 (30 day dosing starting day 1) in CT26 tumor model;
  • FIG. 9 is a line graph showing that 25 (4 days starting on day 6) is superior to CB-839 (continuous twice daily dosing starting on day 1 prior to engraftment) in a CT26 tumor model.
  • FIG. 9 shows mice received daily 25 (1.9 mg/kg) on days 6-9 vs BID glutaminase inhibitor on days 1-15;
  • FIG. 10 is a line graph showing that 25 (daily days 7-22) is superior to CB-839 (continuous twice daily dosing days 1-29) in a 4T1 breast cancer model. Mice received daily 25 (1.0 mg ⁇ kg/d) for days 7-22 as compared to BID glutaminase inhibitor for days 1-29;
  • FIGS. 11A-D and FIG. 11F are line graphs, and FIG. 11E is a Kaplan-Meier graph showing that 25 dosing of 1 mg/kg following by 0.3 mg/kg leads to a complete and durable response in the MC38 tumor;
  • FIG. 12A , FIG. 12B , and FIG. 12D are line graphs, and FIG. 12C is a Kaplan-Meier graph showing that 25 gives a robust response and improved overall survival in multiple tumor models including, for example, CT26 Colon Cancer;
  • FIG. 13A , FIG. 13B , and FIG. 13D are line graphs, and FIG. 13C is a bar graph showing that 25 provides a robust response and improved overall survival in multiple tumor models including, for example, 4T1 breast cancer;
  • FIGS. 14A-G are line graphs showing that mice cured with 25 alone immunologically reject tumors upon re-challenge, demonstrating that 25 monotherapy is immunotherapy;
  • FIG. 15A and FIG. 15C are line graphs and FIG. 15B and FIG. 15D are Kaplan-Meier graphs showing that 25 monotherapy is immunotherapy;
  • FIGS. 16A-F are line graphs showing that glutamine inhibition (e.g., DON) reduces the oxygen consumption and lactate production of tumor cells;
  • glutamine inhibition e.g., DON
  • FIGS. 17A-F are graphs showing that glutamine inhibition (e.g., DON) improved the CD8/Treg ratio in the tumor and reduces hypoxia in the TILs;
  • glutamine inhibition e.g., DON
  • FIGS. 18A-D are line graphs and FIG. 18E is a Kaplan-Meier graph showing that 25 conditions the tumor to be eliminated by anti-PD1 therapy in the MC38 Model, and in particular that 25 unexpectedly rescues anti-PD1 failures;
  • FIGS. 19A-C are line graphs and FIG. 19D is a Kaplan-Meier graph showing that even in the more difficult CT26 model, 25 enhances the response to anti-PD1 therapy;
  • FIG. 20A is a line graph
  • FIG. 20B is a Kaplan-Meier graph showing that inhibiting glutamine metabolism also unexpectedly potentiates the anti-tumor response to adenosine A2a receptor (A2aR) blockade;
  • A2aR adenosine A2a receptor
  • FIG. 21A is a line graph
  • FIG. 21B is an illustration
  • FIG. 21C is a Kaplan-Meier graph showing that inhibiting glutamine metabolism unexpectedly enhances the efficacy of adoptive cellular therapy (ACT) in a B16-OVA model;
  • ACT adoptive cellular therapy
  • FIG. 22A is a line graph
  • FIG. 22B is a bar graph
  • FIG. 22C is a table showing the in vivo pharmacokinetics of DON following i.v. administration of DON (1) and 14b in monkey plasma and cerebrospinal fluid (CSF).
  • 1 and 14b were dosed in two pigtail macaques at 1.6 mg/kg equivalent of 1 via i.v. administration and plasma (0.25-6 h) and CSF (30 min) concentrations of DON were evaluated via LC/MS.
  • Relative to 1 14b delivered substantially lower DON plasma concentration.
  • the reverse was observed in CSF, where 14b delivered significantly higher DON CSF concentrations, achieving an unexpected 10-fold enhanced CSF to plasma ratio at 30 minute post dose;
  • FIG. 23 is a bar graph showing species specific plasma stability of (14b); 14b is stable in plasma of human, pig, dog and monkeys, but rapidly metabolized in mice;
  • FIG. 24 is an illustration showing exemplary structures of DON and DON-based prodrugs 25, 9, 38, 38a, 60, and 60a; different N-amino acid promoeities (e,g, leucine, tryptophan) provide differential plasmas and microsomal stability;
  • N-amino acid promoeities e,g, leucine, tryptophan
  • FIG. 25A , FIG. 25B , FIG. 25C , and FIG. 25D are bar graphs showing the in vitro plasma stability of DON prodrugs 25, 9, 38a and 60a. Metabolism occurs via removal of N-protecting group; both ethyl and isopropyl esters are stable in plasma of pigs and humans;
  • FIG. 26A , FIG. 26B , FIG. 26C , and FIG. 26D are bar graphs showing the in vitro liver microsomal stability of DON prodrugs 25, 9, 38a and 60a; all prodrugs showed moderate-high stability in human and pig microsomes;
  • FIG. 27A , FIG. 27B , FIG. 27C , FIG. 27D , FIG. 27E , FIG. 27F , FIG. 27G , FIG. 27H , FIG. 27I , and FIG. 27J are bar graphs showing the results of ex-vivo studies in whole human and pig blood of 25, 9, 38a and 60a; DON prodrugs selectively deliver DON to PBMCs in both humans and pigs vs plasma; compared to DON, the PBMC/plasma ratio was unexpectedly enhanced 10-100+ fold;
  • FIG. 28A FIG. 28B , FIG. 28C , FIG. 28D and FIG. 28E are line graphs showing the results of pig in vivo studies with DON prodrugs of 25, 9, 38a and 60a; DON prodrugs selectively deliver DON to PBMCs vs plasma; compared to DON, the PBMC/plasma ratio was expectedly enhanced 6- to 10-fold;
  • FIG. 29A , FIG. 29B , and FIG. 29C are bar graphs showing the plasma stability of compound Methyl-POM 14b and its derivatives
  • FIG. 30 is an illustration showing exemplary structures of N-acylalkyloxy DON-based prodrug analogs for intracellular targeting and brain penetration; the addition of steric bulk to the “bridge” might result in a slower hydrolysis;
  • FIG. 31 is a line graph showing that anti-PD1 monotherapy does not work in a 4T1 tumor model.
  • 4T1 tumor cells (0.1 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice.
  • Anti-PD1 (5 mg/Kg) was administered on day 3, 5, 8, and 11 and tumor volume was measured 2-3 times weekly until tumors were evaluated for tumor infiltrating cells.
  • Group: a-PD1 (d3, 5, 8, 11 100 ug/mouse) 5 mg/kg I.P.;
  • FIG. 32A is an illustration of mice and FIG. 32B is a line graph illustrating that compound 25 inhibits tumor growth.
  • FIG. 32A shows mice after 30 days inoculation (8 days drug free).
  • FIG. 32B shows tumor area vs. days post injection with 4T1 tumor cells.
  • Mice were treated with compound 25 1 mg/kg every day (from d5-d22) or vehicle.
  • 4T1 tumor cells (0.1 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice.
  • Mice received vehicle (PBS) or 1 mg/kg compound 25 daily from day 5 to day 22. Photos were taken on day 30 after tumor inoculation ( FIG. 57A ).
  • Tumor volume was measured 2-3 times weekly until WT mice were sacrificed (when size reached to 20 mm length or necrosis occurred).
  • Day 0 100K 4T1 cells s.c. in 4th mammary pad.
  • Day 5-22 Daily compound 25. The results show that compound 25 inhibited the growth of mammary carcinoma tumor cells.
  • FIG. 33A is an illustration of mice and FIG. 33B is a bar graph illustrating that compound 25 inhibits tumor growth.
  • 4T1-Luc tumor cells (0.1 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice. Mice received vehicle (PBS) or 1 mg/kg compound 25 daily from day 7.
  • Anti-PD1 antibody (5 mg/Kg) was administered on day 5, 8, and 12.
  • Mice carrying 4T1-luc tumors are injected with Luciferin to measure luminescence from the tumor. IVIS imaging were taken on day 13. The results unexpectedly show that treatment with compound 25 as a single agent inhibited mammary carcinoma cell proliferation better than anti-PD1.
  • FIG. 34A is a line graph and FIG. 34B is a bar graph showing that compound 25 inhibits tumor growth.
  • FIG. 34B shows tumor weight (mg) on harvest day 21.
  • 4T1-Luc tumor cells (0.1 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice. Mice received vehicle (PBS) or 1 mg/kg 25 daily from day 7 to day 16.
  • Anti-PD1 antibody (5 mg/Kg) was administered on day 5, 8, 12 and 17. Tumor volume was measured 2-3 times weekly until tumors were evaluated for tumor infiltrating cells on day 21. Tumor weights were measured on day 21.
  • the PD1 group tumor size looks like it was reduced, however, the result was not due to a tumor size reduction, but rather because one of the large tumor mice died;
  • FIGS. 35A-C are line graphs showing reduced CD11b+ cells and G-MDSCs in 25 treated mice, demonstrating that metabolic reprogramming agents that decrease glutamine metabolic activity inhibit myeloid derived suppressor cells.
  • 4T1-Luc tumor cells (0.1 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice. Mice received vehicle (PBS) or 1 mg/kg 25 daily from day 7 to day 16. Anti-PD1 (5 mg/Kg) was administered on day 5, 8, 12 and 17. Percentages of myeloid-derived suppressor cell (MDSC) from circulating blood were monitored on day 6, 9 and 17 by flow cytometry with CD11b and Ly6C/Ly6G.
  • Granulocytic MDSC (G-MDSC): CD11b+Ly6 g+Ly6c lo
  • Monocytic MDSC (Mo-MDSC): CD11b+Ly6G-Ly6c Hi;
  • FIG. 36 is a graph showing increased TNF alpha in 25 treated mice, demonstrating that metabolic reprogramming agents that decrease glutamine metabolic activity increase inflammatory tumor associated macrophages (TAM).
  • 4T1-Luc tumor cells (0.2 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice. Mice received vehicle (PBS) or 1 mg/kg 25 daily from day 7 to day 16. Anti-PD1 (5 mg/Kg) was administered on day 5, 8, 12 and 17. Tumors were evaluated for tumor infiltrating cells on day 21. Cells were seeded on plates and golgi plug 200 ul were added to inhibit cytokine secretion for over night (no additional stimulation).
  • Tumor associated macrophages markers Live/CD45+/CD11b+/F4-80+/CD8 ⁇ for flow cytometry analysis. Macrophages derived from tumor. The cells were incubated with Golgi plug o.n. w/o stim. TAM: Live/CD45+/CD11b+/F4-80+/CD8-;
  • FIG. 37A is an illustration
  • FIG. 37B is a graph
  • FIG. 37C is an illustration showing reduced fibrocytes in 25 treated mice, demonstrating that metabolic reprogramming agents that decrease glutamine metabolic activity inhibit bone marrow derived fibrocytes which are thought to play a role in inhibiting immunotherapy, as well as generating an extracellular matrix that surrounds tumors and inhibits chemotherapy.
  • 4T1-Luc tumor cells (0.1 million) were injected into the mammary fat pads of 8-week-old female BALB/c mice. Mice received vehicle (PBS) or 1 mg/kg 25 daily from day 7 to day 16.
  • Anti-PD1 (5 mg/Kg) was administered on day 5, 8, 12 and 17. Tumors were evaluated for tumor infiltrating cells on day 21.
  • Fibrocytes markers: CollagenI+CD11b+CD45+live were used for flow cytometry analysis.
  • FIG. 38A is a line graph demonstrating different DON plasma profiles in Monkey for DON and compound 14b.
  • FIG. 38B is a bar graph showing that compound 14b exhibited enhanced CSF:plasma ratio of DON in Monkey.
  • FIG. 39A is a line graph demonstrating different DON plasma profiles in swine for DON, compound 14b and compound 47.
  • FIG. 39B is a bar graph showing that compounds 14b and 47 exhibited enhanced CSF delivery of DON at 60 min post-administration in swine
  • FIG. 39C is a bar graph showing that compounds 14b and 47 exhibited enhanced CSF:plasma ratio of DON at 60 min post-administration in swine.
  • the presently disclosed subject matter demonstrates that certain conditions, diseases, and/or disorders involve metabolically reprogrammed cells whose activation, function, growth, proliferation, and/or survival in an abnormal, harmful, and/or unhealthy state depend on increased activity of at least one, at least two, or at least three metabolic pathways selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis.
  • the abnormal, harmful, and/or unhealthy state of the cell refers to its effect on or relative to the subject whose cells are affected by the condition, disease, or disorder rather than on or relative to the cell itself which exhibits an increased ability to thrive in the abnormal, harmful, and/or unhealthy state in a manner that is believed to be proportionate to the increase in the activity of at least one, at least two, or at least three metabolic pathways (e.g., glutamine metabolism, glycolysis, and/or fatty acid synthesis).
  • at least one, at least two, or at least three metabolic pathways e.g., glutamine metabolism, glycolysis, and/or fatty acid synthesis.
  • metabolic reprogramming disorders are amenable to treatment using at least one, at least two, or at least three metabolic reprogramming agents that decrease activity of at least one, at least two, or at least three metabolic pathways selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis.
  • the metabolic reprogramming disorders comprise conditions, diseases, or disorders that involve aberrant and/or excessive glutamine metabolism, aberrant and/or excessive glycolysis, or aberrant and/or excessive fatty acid synthesis.
  • the term “excessive glutamine metabolism” means an increase in the amount of glutamine metabolic activity in a subject with a condition, disease, or disorder (e.g., a metabolic reprogramming disorder) as compared to the amount of glutamine metabolic activity in a subject without a similar disease or condition, such as an increase of approximately 100%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, or more.
  • the term “aberrant glutamine metabolism” means a change in the biological activity of glutamine in a subject with a condition, disease or disorder (e.g., a metabolic reprogramming disorder) as compared to the glutamine activity in a subject without a similar condition, disease, or disorder, such as increased utilization of glutamine in the growth and/or proliferation of malignant, neoplastic, or other pathologic cellular processes (e.g., immune disorders, neurodegenerative disorders, inflammatory disorders, etc.).
  • a condition, disease or disorder e.g., a metabolic reprogramming disorder
  • pathologic cellular processes e.g., immune disorders, neurodegenerative disorders, inflammatory disorders, etc.
  • the term “excessive glycolysis metabolism” means an increase in the amount of glycolytic metabolic activity in a subject with a condition, disease, or disorder (e.g., a metabolic reprogramming disorder) as compared to the amount of glycolytic metabolic activity in a subject without a similar disease or condition, such as an increase of approximately 100%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, or more.
  • the term “aberrant glycolytic metabolism” means a change in the biological activity of glycolysis in a subject with a condition, disease or disorder (e.g., a metabolic reprogramming disorder) as compared to the glycolytic activity in a subject without a similar condition, disease, or disorder, such as increased utilization of glucose in the growth and/or proliferation of malignant, neoplastic, or other pathologic cellular processes (e.g., immune disorders, neurodegenerative disorders, inflammatory disorders, etc.).
  • a condition, disease or disorder e.g., a metabolic reprogramming disorder
  • pathologic cellular processes e.g., immune disorders, neurodegenerative disorders, inflammatory disorders, etc.
  • the term “excessive fatty acid synthesis” means an increase in the amount of fatty acid synthesis in a subject with a condition, disease, or disorder (e.g., a metabolic reprogramming disorder) as compared to the amount of fatty acid synthesis in a subject without a similar condition, disease, or disorder, such as an increase of approximately 100%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, or more.
  • the term “aberrant fatty acid synthesis” means a change in the biological activity of fatty acid synthesis in a subject with a condition, disease or disorder (e.g., a metabolic reprogramming disorder) as compared to the fatty acid synthesis in a subject without a similar condition, disease, or disorder, such as increased utilization of fatty acids in the growth and/or proliferation of malignant, neoplastic, or other pathologic cellular processes (e.g., immune disorders, neurodegenerative disorders, inflammatory disorders, etc.).
  • a condition, disease or disorder e.g., a metabolic reprogramming disorder
  • pathologic cellular processes e.g., immune disorders, neurodegenerative disorders, inflammatory disorders, etc.
  • a “metabolically reprogrammed” cell refers to a cell in which the activity of at least one, at least two, or at least three metabolic pathways (e.g., glutamine metabolism, glycolysis, and fatty acid synthesis) has increased in response to the cells energetic and biosynthetic demands placed on the cell in order for the cell to become activated, function, grow, proliferate, and/or survive in the abnormal, harmful, and/or unhealthy state.
  • metabolic pathways e.g., glutamine metabolism, glycolysis, and fatty acid synthesis
  • a “metabolic reprogramming agent” refers to an agent that is capable of reversing the metabolic reprogramming of a cell from a cell whose activation, function, growth, proliferation, and/or survival in an abnormal, harmful, and/or unhealthy state depends on increased activity of at least one, at least two, or at least three metabolic pathways (e.g., glutamine metabolism, glycolysis, and fatty acid synthesis) to a cell that has a decreased capacity or has lost its ability to thrive (e.g., activate, function, grow, proliferate, and/or survive) in the abnormal, harmful and/or unhealthy state.
  • a “metabolic reprogramming agent” inhibits at least one of, at least two of, or all of aberrant and/or excessive glutamine metabolism, aberrant and/or excessive glycolysis, and aberrant and/or excessive fatty acid synthesis.
  • the presently disclosed subject matter provides a method for treating a subject having a condition, disease, or disorder that involves metabolically reprogrammed cells whose activation, function, growth, proliferation, and/or survival depends on increased activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis, the method comprising administering to the subject at least one metabolic reprogramming agent that decreases activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis in an amount effective to treat the condition, disease, or disorder.
  • the presently disclosed subject matter provides a method for treating a subject having a condition, disease, or disorder that involves at least one of aberrant and/or excessive glutamine metabolism, aberrant and/or excessive glycolysis, or aberrant and/or excessive fatty acid synthesis, the method comprising administering to the subject at least one metabolic reprogramming agent that decreases activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis in an amount effective to treat the condition, disease, or disorder.
  • the presently disclosed methods result in a decrease in the severity of a condition, disease, or disorder (e.g., a metabolic reprogramming disorder) in a subject.
  • a condition, disease, or disorder e.g., a metabolic reprogramming disorder
  • the term “decrease” is meant to inhibit, suppress, attenuate, diminish, arrest, or stabilize a symptom of the condition, disease, or disorder.
  • the terms “treat,” “treating,” “treatment,” and the like refer to reducing or ameliorating a disease or condition, and/or symptoms associated therewith. It will be appreciated that, although not precluded, treating a disease or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated.
  • the method comprises administering to the subject at least two metabolic reprogramming agents that decrease the activity of at least two metabolic pathways selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis in an amount effective to treat the condition, disease, or disorder.
  • the method comprises administering to the subject at least three metabolic reprogramming agents that each decrease the activity of a different metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis in an amount effective to treat the condition, disease, or disorder.
  • a “subject” and “patient” are used interchangeably herein.
  • the subject treated by the presently disclosed methods, uses, metabolic reprogramming agents and compositions comprising those agents in their many embodiments is desirably a human subject, although it is to be understood that the methods described herein are effective with respect to all vertebrate species, which are intended to be included in the term “subject.”
  • a “subject” can include a human subject for medical purposes, such as for the treatment of an existing condition or disease or the prophylactic treatment for preventing the onset of a condition or disease, or an animal subject for medical, veterinary purposes, or developmental purposes.
  • Suitable animal subjects include mammals including, but not limited to, primates, e.g., humans, monkeys, apes, and the like; bovines, e.g., cattle, oxen, and the like; ovines, e.g., sheep and the like; caprines, e.g., goats and the like; porcines, e.g., pigs, hogs, and the like; equines, e.g., horses, donkeys, zebras, and the like; felines, including wild and domestic cats; canines, including dogs; lagomorphs, including rabbits, hares, and the like; and rodents, including mice, rats, and the like.
  • mammals including, but not limited to, primates, e.g., humans, monkeys, apes, and the like; bovines, e.g., cattle, oxen, and the like; ovines, e.g., sheep and the like; cap
  • An animal may be a transgenic animal.
  • the subject is a human including, but not limited to, fetal, neonatal, infant, juvenile, and adult subjects.
  • a “subject” can include a patient afflicted with or suspected of being afflicted with a condition or disease.
  • aspects of the invention involve the use of at least one, at least two, or at least three metabolic reprogramming agents, alone, or optionally together in combination with a chemotherapeutic agent, an immunotherapeutic agent, and/or a radiotherapeutic agent, for the treatment of a cancer.
  • the condition, disease, or disorder is a cancer.
  • the metabolically reprogrammed cells comprise malignant or cancerous cells.
  • malignant or cancer cells whose activation, function, growth, proliferation, and/or survival in an abnormal, harmful, or unhealthy state depends on increased metabolic activity of at least one, at least two, or at least three metabolic pathways selected from the group consisting of glutamine metabolism, glycolysis and fatty acid synthesis include, but are not limited to, cMyc-dependent cancer cells, glutamine-dependent cancer cells, and combinations thereof.
  • a “glutamine-dependent cancer cell” is a cancer cell in which glutamine is an important fuel source for cellular energy in the cancer cell (e.g., hematopoietic tumors, hepatomas, Ehrilich carcinoma (see Huber et al., “Uptake of glutamine antimetabolites 6-diazo-5-oxo-L-norleucine (DON) and acivicin in sensitive and resistant tumor cell lines,” Int. J. Cancer. 1988; 41:752-755)).
  • cMyc-dependent cancer cells refers to cancer cells exhibiting activation, overexpression and/or amplification of c-Myc.
  • a “Myc-dependent cancer” is a cancer in which c-Myc plays a role in increased glutamine metabolism in the cancer cells, i.e., cMyc-dependent glutamine addicted cancer cells.
  • Examples of Myc-dependent cancers include, without limitation, lymphoma, neuroblastoma, and small cell lung cancer.
  • cancer maintenance therapy refers to a therapy administered to a cancer patient who is in cancer remission.
  • the presently disclosed subject matter provides a method of preventing a relapse or reducing the incidence of relapse of a cancer subject in remission, the method comprising administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • a metabolic reprogramming agent is a compound having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.
  • “remission” includes partial and complete remission and refers to a decrease in or disappearance of signs and symptoms of cancer.
  • “Partial remission” means that the cancer responded to treatment with the primary therapy, but at least a portion of the tumor and/or at least a portion of the cancerous cells are still present in the subject, for example, at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 49% of a measurable tumor and/or measurable cancerous cells are still present in the subject post-therapy.
  • “Complete remission” means that the subject shows no signs or symptoms of cancer, for example, after a healthcare provider has used the most accurate and up-to-date tests available to detect the cancer and is unable to detect any signs or symptoms of cancer. It is to be understood that cancerous cells might still exist in a subject in complete remission at levels that are undetectable.
  • the metabolic reprogramming agent is administered to the subject post transplant.
  • post transplant refers to a subject that has recently received a cell, tissue, or organ transplantation, including for example, subjects receiving immunosuppressive agents to prevent, or reduce the risk and/or severity of, a transplant rejection.
  • the metabolic reprogramming agent is administered to the subject post chemotherapy.
  • the metabolic reprogramming agent is administered to the subject post immunotherapy.
  • the metabolic reprogramming agent is administered to the subject post photodynamic therapy.
  • the metabolic reprogramming agent is administered to the subject post proton therapy.
  • the metabolic reprogramming agent is administered to the subject post radiotherapy.
  • the metabolic reprogramming agent is administered to the subject post surgery; and combinations thereof. In some embodiments, the metabolic reprogramming agent is administered to the subject at two or more of post transplant, post chemotherapy, post immunotherapy, post photodynamic therapy, post proton therapy, post radiotherapy, post surgery, and combinations thereof.
  • a “cancer” in a subject refers to the presence of cells possessing characteristics typical of cancer-causing cells, for example, uncontrolled proliferation, loss of specialized functions, immortality, significant metastatic potential, significant increase in anti-apoptotic activity, rapid growth and proliferation rate, and certain characteristic morphology and cellular markers.
  • cancer cells will be in the form of a tumor; such cells may exist locally within an animal, or circulate in the blood stream as independent cells, for example, leukemic cells.
  • a “tumor,” as used herein, refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all precancerous and cancerous cells and tissues.
  • a “solid tumor”, as used herein, is an abnormal mass of tissue that generally does not contain cysts or liquid areas.
  • a solid tumor may be in the brain, colon, breasts, prostate, liver, kidneys, lungs, esophagus, head and neck, ovaries, cervix, stomach, colon, rectum, bladder, uterus, testes, and pancreas, as non-limiting examples.
  • the solid tumor regresses or its growth is slowed or arrested after the solid tumor is treated with the presently disclosed methods.
  • the solid tumor is malignant.
  • the cancer comprises Stage 0 cancer.
  • the cancer comprises Stage I cancer.
  • the cancer comprises Stage II cancer.
  • the cancer comprises Stage III cancer. In some embodiments, the cancer comprises Stage IV cancer. In some embodiments, the cancer is refractory and/or metastatic. For example, the cancer may be refractory to treatment with radiotherapy, chemotherapy or monotreatment with immunotherapy.
  • the cancer is a cancer of the central nervous system (CNS cancer). It is believed that certain of the presently disclosed metabolic reprogramming agents and compositions are particularly useful in the treatment of CNS cancers and cancers of CNS origin.
  • CNS cancer central nervous system
  • metabolic reprogramming agents e.g., prodrugs of glutamine analogs, e.g., DON prodrugs, e.g., a compound having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.
  • DON prodrugs e.g., a compound having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.
  • certain of the presently disclosed metabolic reprogramming agents are contemplated for use as cancer therapy (e.g., maintenance therapy), immunotherapy, and an enhancement to immunotherapy, for the treatment of CNS cancers and cancers of CNS origin.
  • Exemplary CNS cancers treatable with the presently disclosed methods, compositions and agents include, without limitation, gliomas, astrocytomas, oligodendrogliomas, ependymoas, mixed gliomas (e.g., oligoastrocytomas), meningiomas (e.g., atypical, invasive, anaplastic, etc.), medulloblastomas, gangliogliomas, schwannomas (neuroliemmomas), craniopharyngiomas, chordomas, non-Hodgkin lymphoma of CNS origin, and, pituitary tumors.
  • the CNS cancer comprises glioblastoma multiform (GBM).
  • the cancer is a cancer that is associated with transplant and/or immunosuppression.
  • organ transplants e.g., kidney, liver, heart, lung etc.
  • the cancer risk is elevated for infection-related cancer due to immunosuppression, for example, because of medications administered to suppress the immune system and prevent transplant rejection (e.g., organ).
  • the cancer associated with transplant and/or immunosuppression is related to an infectious agent.
  • cancers associated with transplant and/or immunosuppression include, without limitation, anal cancer, Kaposi sarcoma, kidney cancer, liver cancer, lung cancer, melanoma, non-Hodgkin's lymphoma, and thyroid cancer.
  • the subject is a child or elderly adult transplant recipient (e.g., liver, heart, kidney, etc.) who may or may not be infected with Epstein-Barr virus.
  • the cancer is a cancer that is refractory to chemotherapy.
  • the cancer is a cancer that is refractory to photodynamic therapy.
  • the cancer is a cancer that is refractory to proton therapy.
  • the cancer is a cancer that is refractory to radiotherapy.
  • the cancer is a cancer that is refractory to surgery.
  • Cancer as used herein includes newly diagnosed or recurrent and/or refractory cancers, including without limitation, acute lymphoblastic leukemia, acute myelogenous leukemia, advanced soft tissue sarcoma, brain cancer, metastatic or aggressive breast cancer, breast carcinoma, bronchogenic carcinoma, choriocarcinoma, chronic myelocytic leukemia, colon carcinoma, colorectal carcinoma, Ewing's sarcoma, gastrointestinal tract carcinoma, glioma, glioblastoma multiforme, head and neck squamous cell carcinoma, hepatocellular carcinoma, Hodgkin's disease, intracranial ependymoblastoma, large bowel cancer, leukemia, liver cancer, lung carcinoma, Lewis lung carcinoma, lymphoma, malignant fibrous histiocytoma, a mammary tumor, melanoma, mesothelioma, neuroblastoma, osteosarcoma, ovarian cancer, pancreatic cancer,
  • the cancer treated is a newly diagnosed, or recurrent, and/or refractory cancer selected from the group consisting of celnasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T-cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger-Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter cancer, brain cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer, osteochondroma, chondrosarcoma, Ewing's sar
  • the condition, disease, or disorder is lymphoma. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of lymphoma in a subject in need thereof, the method comprising administering to the subject at least one metabolic reprogramming agent that decreases glutamine metabolism in an amount effective to treat lymphoma in the subject.
  • the condition, disease, or disorder is melanoma. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of melanoma in a subject in need thereof, the method comprising administering to the subject at least one metabolic reprogramming agent that decreases glutamine metabolism in an amount effective to treat melanoma in the subject.
  • the methods include administering to the subject an effective amount of radiotherapy. In some embodiments, the methods include administering of the subject an effective amount of immunotherapy (e.g., a second immunotherapy). In some embodiments, the methods include administering to the subject an effective amount of photodynamic therapy. In some embodiments, the methods include administering to the subject an effective amount of proton therapy. In some embodiments, the methods include surgically resecting at least a portion of a tumor before, during, or after treatment with the at least one, at least two, or at least three metabolic reprogramming agents, and optionally at least one chemotherapeutic agent, immunotherapeutic agent, and/or radiotherapeutic agent.
  • the condition, disease, or disorder is a newly diagnosed, or recurrent and/or refractory cancer selected from the group consisting of acute lymphoblastic leukemia, acute myelogenous leukemia, advanced soft tissue sarcoma, brain cancer, metastatic or aggressive breast cancer, breast carcinoma, bronchogenic carcinoma, choriocarcinoma, chronic myelocytic leukemia, colon carcinoma, colorectal carcinoma, Ewing's sarcoma, gastrointestinal tract carcinoma, glioma, glioblastoma multiforme, head and neck squamous cell carcinoma, hepatocellular carcinoma, Hodgkin's disease, intracranial ependymoblastoma, large bowel cancer, leukemia, liver cancer, lung carcinoma, Lewis lung carcinoma, lymphoma, malignant fibrous histiocytoma, a mammary tumor, melanoma, mesothelioma, neuroblastoma, osteosarcoma,
  • the cancer is not acute lymphoblastic leukemia. In some embodiments, the cancer is not acute myelogenous leukemia. In some embodiments, the cancer is not advanced soft tissue sarcoma. In some embodiments, the cancer is not brain cancer. In some embodiments, the cancer is not metastatic or aggressive breast cancer. In some embodiments, the cancer is not breast carcinoma. In some embodiments, the cancer is not bronchogenic carcinoma. In some embodiments, the cancer is not choriocarcinoma. In some embodiments, the cancer is not chronic myelocytic leukemia. In some embodiments, the cancer is not colon carcinoma. In some embodiments, the cancer is not colorectal carcinoma. In some embodiments, the cancer is not Ewing's sarcoma.
  • the cancer is not gastrointestinal tract carcinoma. In some embodiments, the cancer is not glioma. In some embodiments, the cancer is not glioblastoma multiforme. In some embodiments, the cancer is not head and neck squamous cell carcinoma. In some embodiments, the cancer is not hepatocellular carcinoma. In some embodiments, the cancer is not Hodgkin's disease. In some embodiments, the cancer is not intracranial ependymoblastoma. In some embodiments, the cancer is not large bowel cancer. In some embodiments, the cancer is not leukemia. In some embodiments, the cancer is not liver cancer. In some embodiments, the cancer is not lung carcinoma. In some embodiments, the cancer is not Lewis lung carcinoma.
  • the cancer is not lymphoma. In some embodiments, the cancer is not malignant fibrous histiocytoma. In some embodiments, the cancer is not a mammary tumor. In some embodiments, the cancer is not melanoma. In some embodiments, the cancer is not mesothelioma. In some embodiments, the cancer is not neuroblastoma. In some embodiments, the cancer is not osteosarcoma. In some embodiments, the cancer is not ovarian cancer. In some embodiments, the cancer is not pancreatic cancer. In some embodiments, the cancer is not a pontine tumor. In some embodiments, the cancer is not premenopausal breast cancer. In some embodiments, the cancer is not prostate cancer.
  • the cancer is not rhabdomyosarcoma. In some embodiments, the cancer is not reticulum cell sarcoma. In some embodiments, the cancer is not sarcoma. In some embodiments, the cancer is not small cell lung cancer. In some embodiments, the cancer is not a solid tumor. In some embodiments, the cancer is not stomach cancer. In some embodiments, the cancer is not testicular cancer. In some embodiments, the cancer is not uterine carcinoma.
  • aspects of the presently disclosed subject matter involve the use of at least one, at least two, or at least three metabolic reprogramming agents, alone, or optionally together in combination with an additional immunotherapy (e.g., checkpoint blockade, adoptive cellular therapy (ACT), vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, for the treatment of cancer.
  • additional immunotherapy e.g., checkpoint blockade, adoptive cellular therapy (ACT)
  • vaccines e.g., tumor vaccines
  • passive immunotherapy antibodies for the treatment of cancer.
  • the presently disclosed subject matter provides a method for treating a cancer in a subject in need thereof, the method comprising: (a) administering a therapeutically effective amount of a first immunotherapy to the subject, wherein the first immunotherapy is a metabolic reprogramming agent; and (b) optionally administering a therapeutically effective amount of a second immunotherapy to the subject.
  • the metabolic reprogramming agent is a glutamine antagonist.
  • the metabolic reprogramming agent is a glutamine analog that interferes with a glutamine metabolic pathway.
  • the metabolic reprogramming agent is selected from the group consisting of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV).
  • the metabolic reprogramming agent is a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway.
  • at least one metabolic reprogramming agent is a prodrug of acivicin, azaserine, DON, and L-DONV.
  • a prodrug of a glutamine antagonist or a pharmaceutically acceptable salt or ester thereof has a structure of formula (I):
  • X is selected from the group consisting of a bond, —O—, and —(CH 2 ) n —, wherein n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • R 1 is selected from the group consisting of H and a first prodrug-forming moiety capable of forming a salt or an ester; and
  • R 2 is H or a second prodrug-forming moiety capable of forming an amide linkage, a carbamate linkage, a phosphoramidate linkage or a phosphorodiamidate linkage with the nitrogen adjacent to R 2 ;
  • R 2′ is selected from the group consisting of H, C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, or R 2 and R 2′ together form a ring structure comprising —C( ⁇ O)-G-C( ⁇ O)—, wherein G is selected from the group consisting of C 1 -C 8 alkylene, C 1 -C 8
  • amide linkage comprises a structure represented by the formula:
  • R v is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heterocyclyl, substituted heterocyclyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkylamine, substituted alkylamine, heteroaryl, and substituted heteroaryl.
  • R w is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heterocyclyl, substituted heterocyclyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkylamine, substituted alkylamine, heteroaryl, and substituted heteroaryl.
  • phosphoramidate linkage comprises a structure represented by the formula:
  • R x and R x′ are each independently selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heterocyclyl, substituted heterocyclyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkylamine, substituted alkylamine, heteroaryl, and substituted heteroaryl.
  • phosphorodiamidate linkage comprises a structure represented by the formula:
  • R y and R z are each independently selected from the group consisting of H, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclyl, substituted heterocyclyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, —(CR 3 R 4 ) m —Z, —(CR 3 R 4 ) m -Q-Z, aryl, substituted aryl, alkylamine, substituted alkylamine, heteroaryl, substituted heteroaryl, and
  • X is —CH 2 —, and n is 1.
  • the prodrug compound has both the first prodrug-forming moiety and the second prodrug-forming moiety.
  • the glutamine analog is a glutamine antagonist, i.e., the prodrug is a prodrug of a glutamine analog that antagonizes a glutamine pathway.
  • glutamine antagonists include, without limitation, 6-diazo-5-oxo-norleucine (DON), and aza-serine, and 5-diazo-4-oxo-L-norvaline (L-DONV).
  • the presently disclosed subject matter provides a prodrug of DON. In some embodiments, the prodrug of DON has a structure of formula (I). In some embodiments, the presently disclosed subject matter provides a prodrug of L-DONV. In some embodiments, the prodrug of L-DONV has a structure of formula (I). In some embodiments, the presently disclosed subject matter provides a prodrug of azaserine. In some embodiments, the prodrug of azaserine has a structure of formula (I).
  • R 1 of formula (I) comprises a residue PRO 1 of the prodrug-forming moiety, which, together with a basic moiety and the terminal hydroxyl group forms a salt.
  • R 1 of formula (I) comprises a residue PRO 1 of the prodrug-forming moiety, which, together with an alkyl group and the oxygen of an adjoining hydroxyl group forms an ester.
  • R 1 of formula (I) comprises a residue PRO 1 of the prodrug-forming moiety, which, together with an alkyl group and the nitrogen adjoining the R 2′ group, forms an azlactone or an oxazolidone.
  • R 1 of formula (I) is selected from the group consisting of H, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkenyl, substituted cycloalkenyl, tri(hydrocarbyl)ammonium, and tetra(hydrocarbyl)ammonium.
  • Preferred alkyl group, cycloalkyl group, alkenyl group, alkynyl group, and cycloalkenyl group substituents include alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl.
  • R 1 of formula (I) is not H. In some embodiments, R 1 of formula (I) is not H when R 2 and R 2′ are H. In some embodiments, R 2 and R 2′ of formula (I) are each H when and R 1 is not H.
  • R 1 of formula (I) is selected from the group consisting of a C 1-6 straight-chain alkyl, a substituted C 1-6 straight-chain alkyl, a C 1-6 branched alkyl, a substituted C 1-6 branched alkyl, tri(C 1 -C 8 -alkyl)ammonium, tetra(C 1 -C 8 -alkyl)ammonium, triphenylammonium, tri(hydroxy-C 1 -C 8 -alkyl)ammonium, and tetra(hydroxy-C 1 -C 8 -alkyl)ammonium.
  • R 1 of formula (I) is selected from the group consisting of methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, trimethylammonium, triethylammonium, tri(hydroxyethyl)ammonium, tripropylammonium, and tri(hydroxypropyl)ammonium.
  • R 1 of formula (I) is methyl.
  • R 1 of formula (I) is ethyl.
  • R 1 of formula (I) is isopropyl.
  • R 2 of formula (I) comprises a residue PRO 2 of the second prodrug-forming moiety, which, together with a carbonyl, oxy carbonyl, or phosphonyl group and the nitrogen of the adjoining NH, forms an amide, a carbamate, phosphoramidate, or phosphorodiamidate linkage.
  • R 2 of formula (I) comprises a moiety selected from the group consisting of an amino acid, an N-substituted amino acid, a peptide, a substituted peptide, a monocyclic ring, a substituted monocyclic ring, a bicyclic ring, a substituted bicyclic ring, a purine nucleoside, a substituted purine nucleoside, a pyrimidine nucleoside, and a substituted pyrimidine nucleoside.
  • R 2 of formula (I) is selected from the group consisting of H, alkyl, —C( ⁇ O)—Ar, —C( ⁇ O)—Y—(CR 3 R 4 ) m —Ar, —C( ⁇ O)—Y—(CR 3 R 4 ) m —NR 5 R 6 , —P( ⁇ O)(OR 7 ) n (NHR 9 ) o , —C( ⁇ O)—Y—(CR 3 R 4 ) m —Ar—O—C( ⁇ O)—R 8 , —C( ⁇ O)—Y—(CR 3 R 4 ) m —Ar—O—R 8 , —C( ⁇ O)—O—(CR 3 R 4 ) m —O—C( ⁇ O)—R 10 , —C( ⁇ O)—O—R 9 , —C( ⁇ O)—Y—(CR 3 R 4 ) m —Ar—O—C( ⁇ O)—R 10
  • each R 5 and R 6 is independently H, alkyl, —C( ⁇ O)—(CR 3 R 4 ) m , —C( ⁇ O)—(NR 5 R 6 ), or —C( ⁇ O)—(CR 3 R 4 ) m —NR 5 R 6 ; each R 7 is independently selected from the group consisting of H, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclyl, substituted heterocyclyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, —(CR 3 R 4 ) m —Z, —(CR 3 R 4 ) m -Q-Z, wherein Q is a monosaccharide, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and wherein Z is
  • each R 9 is independently selected from the group consisting of H, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, heterocyclyl, substituted heterocyclyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, —(CR 3 R 4 ) m —Z, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and
  • R 1 and X are as defined above, provided that R 1 is not H; each R 8 is independently alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, monosaccharide, acylated monosaccharide, aryl, substituted aryl, heteroaryl, substituted heteroaryl; each R 10 is independently alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, monosaccharide, acylated monosaccharide, aryl, substituted aryl, heteroaryl, substituted heteroaryl; and Ar is aryl, substituted aryl, heteroaryl, or substituted heteroaryl. It should be appreciated that in addition to substitutions on the amino group of Z, one or more substitutions R 3 , R 4 , R 5 , and/or R 6 can be made to the 5 or 6 membered rings of Z.
  • Embodiments I-LXI The disclosure also provides the following particular embodiments numbered Embodiments I-LXI.
  • a method of treating cancer in a subject comprising administering to the subject in need thereof a compound, or a pharmaceutically acceptable salt thereof, having formula (I):
  • X is selected from the group consisting of a bond, —O—, and —(CH 2 ) n —, wherein n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • R 1 is selected from the group consisting of C 1-6 alkyl and substituted C 1-6 alkyl;
  • R 2 is an amino acid, an N-substituted amino acid, or —C( ⁇ O)—O—(CR 3 R 4 ) m —O—C( ⁇ O)—R 10 ;
  • R 2′ is selected from the group consisting of H, C 1 -C 6 alkyl, and substituted C 1 -C 6 alkyl; each R 3 and R 4 are independently H, C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, aryl, substituted aryl, —(CR 3 R 4 ) m —NR 5 R 6 , or
  • n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • R 5 and R 6 are independently H or alkyl
  • R 10 is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, monosaccharide, acylated monosaccharide, aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • R 1 is selected from the group consisting of methyl, ethyl, isopropyl, cyclopentyl, cyclohexyl, trimethylammonium, triethylammonium, tri(hydroxyethyl)ammonium, tripropylammonium, and tri(hydroxypropyl)ammonium.
  • R 2 is selected from the consisting of —C( ⁇ O)—Y—(CR 3 R 4 ) m —NR 5 R 6 , and —C( ⁇ O)—O—(CR 3 R 4 ) m —O—C( ⁇ O)—R 10 ; wherein:
  • Y is —O— or a bond
  • n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • each R 3 and R 4 is independently H, C 1 -C 6 alkyl or substituted C 1 -C 6 alkyl, aryl or substituted aryl;
  • R 10 is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • n 1;
  • R 5 and R 6 are each H.
  • R 1 is selected from the group consisting of H and C 1-6 alkyl
  • R 11 is selected from the group consisting of H, methyl, isopropyl, sec-butyl, CH 2 CH(CH 3 ) 2 , benzyl, p-hydroxybenzyl CH 2 OH, CH(OH)CH 3 , CH 2 -3-indoyl, CH 2 COOH, CH 2 CH 2 COOH, —CH 2 C(O)NH 2 , CH 2 CH 2 C(O)NH 2 , CH 2 SH, CH 2 CH 2 SCH 3 , (CH 2 ) 4 NH 2 , (CH 2 ) 3 NHC( ⁇ NH)NH 2 , and CH 2 -3-imidazoyl;
  • R 12 is selected from the group consisting of H, C 1-4 alkyl, and —C( ⁇ O)R 13 ;
  • R 13 is C 1-4 alkyl.
  • R 1 is selected from the group consisting of H and C 1-6 alkyl
  • R 11 is selected from the group consisting of H, methyl, isopropyl, sec-butyl, CH 2 CH(CH 3 ) 2 , benzyl, p-hydroxybenzyl CH 2 OH, CH(OH)CH 3 , CH 2 -3-indoyl, CH 2 COOH, CH 2 CH 2 COOH, —CH 2 C(O)NH 2 , CH 2 CH 2 C(O)NH 2 , CH 2 SH, CH 2 CH 2 SCH 3 , (CH 2 ) 4 NH 2 , (CH 2 ) 3 NHC( ⁇ NH)NH 2 , and CH 2 -3-imidazoyl;
  • R 12 is selected from the group consisting of H, C 1-4 alkyl, and —C( ⁇ O)R 13 ;
  • R 13 is C 1-4 alkyl.
  • R 1 is C 1-4 alkyl
  • R 11 is selected from the group consisting of isopropyl, sec-butyl, CH 2 CH(CH 3 ) 2 , and CH 2 -3-indoyl;
  • R 12 is selected from the group consisting of H and —C( ⁇ O)R 13 ;
  • R 13 is C 1-4 alkyl.
  • R 1 is selected from the group consisting of H and C 1-6 alkyl
  • R 3 and R 4 are independently selected from the group consisting of H, C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, aryl, and substituted aryl;
  • R 10 is C 1-6 alkyl.
  • R 3 is selected from the group consisting of C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, aryl, and substituted aryl;
  • R 4 is H.
  • R 3 is H
  • R 4 is selected from the group consisting of C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, aryl, and substituted aryl.
  • Embodiment XIX wherein the compound selected from the group consisting of:
  • a method of treating cancer in a subject comprising administering to the subject in need thereof a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
  • Embodiment XXII wherein the immunotherapeutic agent is an immune checkpoint blockade therapy.
  • Embodiment XXIII wherein the immune checkpoint blockade therapy is selected from the group consisting of PD-1 antagonists, PD-L1 antagonists, CTLA-4 antagonists, LAG3 antagonists, and B7-H3 antagonists, and combinations thereof.
  • Embodiment XXII The method of Embodiment XXII, wherein the immunotherapeutic agent is an adoptive cellular therapy.
  • Embodiment XXII wherein the immunotherapeutic agent is marrow-infiltrating lymphocytes (MILs).
  • MILs marrow-infiltrating lymphocytes
  • Embodiment XXII wherein the immunotherapeutic agent is an adenosine A2aR inhibitor.
  • Embodiment XXII wherein the immunotherapeutic agent is a tumor vaccine.
  • Embodiment XXII wherein the immunotherapeutic agent is a passive immunotherapy antibody.
  • Embodiment XXIX wherein the passive immunotherapy antibody is selected from the group consisting of bevacizumab, cetuximab, rituximab, trastuzumab, alemtuzumab, ibritumomab tiuxetan, and panitumumab, and combinations thereof.
  • the cancer is a newly diagnosed, recurrent, and/or refractory cancer selected from the group consisting of celnasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T-cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger-Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter cancer, brain cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer, osteochondroma, chondros
  • a method of preventing a relapse or reducing the incidence of relapse of a cancer subject in remission comprising administering to the subject in need thereof a compound, or a pharmaceutically acceptable salt thereof, having formula (I):
  • X is selected from the group consisting of a bond, —O—, and —(CH 2 )—, wherein n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • R 1 is selected from the group consisting of C 1-6 alkyl and substituted C 1-6 alkyl;
  • R 2 is an amino acid, an N-substituted amino acid, or —C( ⁇ O)—O—(CR 3 R 4 ) m —O—C( ⁇ O)—R 10 ;
  • R 2′ is selected from the group consisting of H, C 1 -C 6 alkyl, and substituted C 1 -C 6 alkyl;
  • each R 3 and R 4 are independently H, C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, aryl, substituted aryl, —(CR 3 R 4 ) m —NR 5 R 6 , or
  • n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • R 5 and R 6 are independently H or alkyl
  • R 10 is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, monosaccharide, acylated monosaccharide, aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • Y is —O— or a bond
  • n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8;
  • each R 3 and R 4 is independently H, C 1 -C 6 alkyl or substituted C 1 -C 6 alkyl, aryl or substituted aryl;
  • R 10 is selected from the group consisting of alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • n 1;
  • R 5 and R 6 are each H.
  • Embodiment XLI wherein the amino acid is tryptophan.
  • Embodiment XXXIII or a pharmaceutically acceptable salt thereof, having formula (IIA):
  • R 1 is selected from the group consisting of H and C 1-6 alkyl
  • R 11 is selected from the group consisting of H, methyl, isopropyl, sec-butyl, CH 2 CH(CH 3 ) 2 , benzyl, p-hydroxybenzyl CH 2 OH, CH(OH)CH 3 , CH 2 -3-indoyl, CH 2 COOH, CH 2 CH 2 COOH, —CH 2 C(O)NH 2 , CH 2 CH 2 C(O)NH 2 , CH 2 SH, CH 2 CH 2 SCH 3 , (CH 2 ) 4 NH 2 , (CH 2 ) 3 NHC( ⁇ NH)NH 2 , and CH 2 -3-imidazoyl;
  • R 12 is selected from the group consisting of H, C 1-4 alkyl, and —C( ⁇ O)R 13 ;
  • R 13 is C 1-4 alkyl.
  • R 1 is selected from the group consisting of H and C 1-6 alkyl
  • R 11 is selected from the group consisting of H, methyl, isopropyl, sec-butyl, CH 2 CH(CH 3 ) 2 , benzyl, p-hydroxybenzyl CH 2 OH, CH(OH)CH 3 , CH 2 -3-indoyl, CH 2 COOH, CH 2 CH 2 COOH, —CH 2 C(O)NH 2 , CH 2 CH 2 C(O)NH 2 , CH 2 SH, CH 2 CH 2 SCH 3 , (CH 2 ) 4 NH 2 , (CH 2 ) 3 NHC( ⁇ NH)NH 2 , and CH 2 -3-imidazoyl;
  • R 12 is selected from the group consisting of H, C 1-4 alkyl, and —C( ⁇ O)R 13 ;
  • R 13 is C 1-4 alkyl.
  • R 1 is C 1-4 alkyl
  • R 11 is selected from the group consisting of isopropyl, sec-butyl, CH 2 CH(CH 3 ) 2 , and CH 2 -3-indoyl;
  • R 12 is selected from the group consisting of H and —C( ⁇ O)R 13 ;
  • R 13 is C 1-4 alkyl.
  • R 1 is selected from the group consisting of H and C 1-6 alkyl
  • R 3 and R 4 are independently selected from the group consisting of H, C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, aryl, and substituted aryl;
  • R 10 is C 1-6 alkyl.
  • a method of preventing a relapse or reducing the incidence of relapse of a cancer subject in remission comprising administering to the subject in need thereof a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
  • Embodiment LIII wherein the immunotherapeutic agent is an immune checkpoint blockade therapy.
  • Embodiment LIV wherein the immune checkpoint blockade therapy is selected from the group consisting of PD-1 antagonists, PD-L1 antagonists, CTLA-4 antagonists, LAG3 antagonists, and B7-H3 antagonists, and combinations thereof.
  • Embodiment LIII wherein the immunotherapeutic agent is an adoptive cellular therapy.
  • Embodiment LIII wherein the immunotherapeutic agent is marrow-infiltrating lymphocytes (MILs).
  • MILs marrow-infiltrating lymphocytes
  • Embodiment LIII wherein the immunotherapeutic agent is an adenosine A2aR inhibitor.
  • Embodiment LIII wherein the immunotherapeutic agent is a tumor vaccine.
  • Embodiment LIII wherein the immunotherapeutic agent is a passive immunotherapy antibody.
  • Embodiment LX wherein the passive immunotherapy antibody is selected from the group consisting of bevacizumab, cetuximab, rituximab, trastuzumab, alemtuzumab, ibritumomab tiuxetan, and panitumumab, and combinations thereof.
  • Embodiments 1-20 The disclosure also provides the following particular embodiments numbered Embodiments 1-20.
  • a method for treating a cancer in a subject in need thereof comprising:
  • the metabolic reprogramming agent is selected from the group consisting of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV).
  • the at least one metabolic reprogramming agent is a prodrug of acivicin, azaserine, DON, and L-DONV.
  • Embodiment 1 further comprising simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an immune checkpoint blockade therapy.
  • the immune checkpoint blockade therapy is selected from the group consisting of PD-1 antagonists, PD-L1 antagonists, CTLA-4 antagonists, LAG3 antagonists, B7-H3 antagonists, and combinations thereof.
  • Embodiment 1 The method of Embodiment 1, further comprising simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an adoptive cellular therapy.
  • Embodiment 1 further comprising simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is marrow-infiltrating lymphocytes (MILs).
  • MILs marrow-infiltrating lymphocytes
  • Embodiment 1 further comprising simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an adenosine A2aR blockade.
  • Embodiment 1 The method of Embodiment 1, further comprising simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a tumor vaccine.
  • Embodiment 1 further comprising simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a passive immunotherapy antibody.
  • Embodiment 14 wherein the passive immunotherapy antibody is selected from the group consisting of bevacizumab, cetuximab, rituximab, trastuzumab, alemtuzumab, ibritumomab tiuxetan, panitumumab, and combinations thereof.
  • Embodiment 1 further comprising simultaneously or sequentially administering to the subject a therapeutically effective amount of a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and
  • Embodiment 1 wherein the first immunotherapy, and the second immunotherapy if administered, is/are administered to the subject in the absence of a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and
  • the cancer is a newly diagnosed, recurrent, and/or refractory cancer selected from the group consisting of celnasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T-cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger-Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer, osteochondroma, chondrosarcoma, Ewing's sar
  • a method of preventing a relapse or reducing the incidence of relapse of a cancer subject in remission comprising administering to the subject a therapeutically effective amount of a metabolic reprogramming agent, wherein the metabolic reprogramming agent is selected from the group consisting of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV), and prodrugs thereof.
  • acivicin L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid
  • azaserine and 6-diazo-5-oxo-norleucine (DON)
  • DON 6-diazo-4-oxo-
  • a diastereomeric mixture of isopropyl (2S)-6-diazo-5-oxo-2-(((1-(pivaloyloxy)ethoxy)carbonyl)amino)hexanoate was prepared and separated by column chromatography to give isopropyl (S)-6-diazo-5-oxo-2-((((S)-1-(pivaloyloxy)ethoxy)carbonyl)amino)hexanoate and isopropyl (S)-6-diazo-5-oxo-2-(((R)-1-(pivaloyloxy)ethoxy)carbonyl) amino)hexanoate.
  • the S,S-isomer was arbitrarily designated compound 14a, and the S,R-isomer was arbitrarily designated compound 14b.
  • the actual stereochemistry of the acetal methyl group was not determined.
  • the diastereoisomer that was arbitrarily designated compound 14b was used in the biological studies described herein. See PCT/US2016/044767 (WO 2017/023774 A1), which is fully incorporated by reference herein.
  • aspects of the presently disclosed subject matter involve the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in a combination immunotherapy together with checkpoint blockade modulators, for example, to enhance checkpoint blockade therapies for the treatment of cancer.
  • the presently disclosed subject matter involves the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in combination immunotherapy together with A2aR blockade, and optionally in combination with a third immunotherapy, a fourth immunotherapy, and/or a fifth immunotherapy, such as tumor vaccines, A2aR blockade, and/or adoptive cell therapy.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an immune checkpoint blockade modulator.
  • the term “immune checkpoint modulator” refers to an agent that totally or partially reduces, inhibits, interferes with, activates, or modulates one or more checkpoint proteins (i.e., an immune checkpoint receptor or a ligand for the immune checkpoint receptor).
  • immune checkpoint modulators of use herein include, but are not limited to, small organic molecules (e.g., haptens) or small inorganic molecules; saccharides; oligosaccharides; polysaccharides; a biological macromolecule selected from the group consisting of peptides (e.g., aptides), proteins, peptide analogs and derivatives; peptidomimetics; nucleic acids selected from the group consisting of miRNAs, siRNAs, shRNAs, antisense nucleic acids, such as antisense RNAs, ribozymes, and aptamers; an extract made from biological materials selected from the group consisting of bacteria, plants, fungi, animal cells, and animal tissues; naturally occurring or synthetic compositions; and any combination thereof.
  • Other examples of immune checkpoint modulators include orthosteric inhibitors, allosteric regulators, interfacial binders, and molecular analogues of substrates that act as competitive inhibitors.
  • immune checkpoint modulators include, without limitation, PD-1 antagonists, PD-L1 antagonists, CTLA-4 antagonists, Lag-3 antagonists, CD137 antagonists, KIR antagonists, Tim3 antagonists, Ox40 agonists, B7-H3 antagonists, and combinations thereof.
  • CTLA-4 antagonists include, without limitation, ipilimumab, tremelimumab and combinations thereof.
  • Anti-CTLA-4 antibodies are currently undergoing clinical trials for the treatment of melanoma.
  • Exemplary Lag-3 antagonists include, without limitation, BMS-986016 and IMP321.
  • CD137 antagonists include, without limitation, CD137-specific antibody, peptide, organic small molecule, antisense oligonucleotide, siRNA, antisense expression vector or recombinant virus.
  • the CD137-specific antibody is clone BBK-2 or clone 4B4-1, as described in WIPO International Application Publication No. WO200405513A2, which is incorporated herein by reference in its entirety.
  • T-cell immunoglobulin and mucin domain 3 (TIM3) antagonists e.g., anti-TIM3 antibodies
  • TIM3 antibodies T-cell immunoglobulin and mucin domain 3
  • exemplary Tim3 antagonists include, without limitation, anti-TIM3 monoclonal antibodies, for example, as described in the poster presentation by Jun et al. “Generation of antagonistic anti-TIM-3 and anti-LAG-3 monoclonal antibodies for potential novel immunotherapy combinations”, available on the world wide web at http://www.tesarobio.com/documents/2014AACRposterLB266.pdf, which is incorporated herein by reference.
  • Ox40 agonists are described by Linch et al., “OX40 Agonists and Combination Immunotherapy: Putting the Pedal to the Metal” Front Oncol 5:34; 2015, which is incorporated herein by reference in its entirety.
  • Exemplary Ox40 agonists include, without limitation, anti-Ox40 agonists antibodies.
  • Other exemplary Ox40 agonists include, without limitation, OX86 and Fc-OX40L.
  • Exemplary B7-H3 antagonists include, without limitation, MGA271.
  • Exemplary PD-L1 antagonists include, without limitation, BMS-936559/MDX-1105, MEDI4736, MPDL3280A, MPDL3280A, MSB0010718C, and combinations thereof.
  • PD-L1 antagonists are currently undergoing clinical trials, for example, for the treatment of melanoma, non-small cell lung cancer, renal cell carcinoma, and ovarian cancer.
  • PD-1 antagonists have been reviewed (see, e.g., Dolan and Gupta 2014).
  • Exemplary PD-1 antagonists of use herein include, without limitation, AMP-224, AMP-554, nivolumab, pembrolizumab, pidilizumab, and combinations thereof.
  • the PD-1 antagonists comprise anti-PD-1 antibodies.
  • anti-PD-1 antibodies include, without limitation, atezolizumab, nivolumab, pembrolizumab, pidilizumab, and combinations thereof.
  • PD-1 antagonists are currently undergoing clinical trials, for example, for the treatment of colorectal cancer, gastric cancer, melanoma, non-small cell lung cancer, ovarian cancer, pancreatic cancer, and renal cell carcinoma.
  • the presently disclosed subject matter provides a method of treating an advanced solid tumor, the method comprising: (a) administering to the subject a therapeutically effective amount of BMS-936559; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating an advanced solid tumor, the method comprising: (a) administering to the subject a therapeutically effective amount of MEDI4736; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating melanoma, the method comprising: (a) administering to the subject a therapeutically effective amount of MPDL3280A in combination with vemurafenib; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating melanoma, the method comprising: (a) administering to the subject a therapeutically effective amount of MEDI4736 in combination with dabrafenib and trametinib; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating melanoma, the method comprising: (a) administering to the subject a therapeutically effective amount of MEDI4736 in combination with trametinib; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating non-small cell lung cancer, the method comprising: (a) administering to the subject a therapeutically effective amount of MPDL3280A; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • MPDL3280A is administered in combination with erlotinib.
  • the presently disclosed subject matter provides a method of treating non-small cell lung cancer, the method comprising: (a) administering to the subject a therapeutically effective amount of MEDI4736; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • MEDI4736 is administered in combination with tremelimumab.
  • the presently disclosed subject matter provides a method of treating renal cell carcinoma, the method comprising: (a) administering to the subject a therapeutically effective amount of MPDL3280A; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • MPDL3280A is administered in combination with bevacizumab.
  • the presently disclosed subject matter provides a method of treating a solid or hematological malignancy, the method comprising: (a) administering to the subject a therapeutically effective amount of MPDL3280A; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating a solid tumor, the method comprising: (a) administering to the subject a therapeutically effective amount of MPDL3280A; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • MPDL3280A is administered in combination with bevacizumab and/or chemotherapy.
  • MPDL3280A is administered in combination with cobimetinib.
  • the presently disclosed subject matter provides a method of treating a solid tumor, the method comprising: (a) administering to the subject a therapeutically effective amount of MEDI4736; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • MEDI4736 is administered in combination with tremelimumab.
  • the presently disclosed subject matter provides a method of treating a solid tumor, the method comprising: (a) administering to the subject a therapeutically effective amount of MSB0010718C; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating advanced cancer, the method comprising: (a) administering to the subject a therapeutically effective amount of AMP-224; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating an advanced solid tumor, the method comprising: (a) administering to the subject a therapeutically effective amount nivolumab in combination with iliolumbar (anti-KIR); and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating a castration-resistant prostate cancer, hepatocellular carcinoma, melanoma, non-small cell lung cancer, or renal cell carcinoma, the method comprising: (a) administering to the subject a therapeutically effective amount of nivolumab; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating colon cancer, gastric cancer, head and neck cancer, Hodgkin lymphoma, melanoma, myeloma, myelodysplastic syndrome, non-Hodgkin lymphoma, non-small cell lung cancer, solid tumors, or triple-negative breast cancer, the method comprising: (a) administering to the subject a therapeutically effective amount of pembrolizumab; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating gastric cancer, pancreatic cancer, small-cell lung cancer, glioblastoma, or triple-negative breast cancer, the method comprising: (a) administering to the subject a therapeutically effective amount of nivolumab in combination with ipilimumab; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating a malignant glioma, the method comprising: (a) administering to the subject a therapeutically effective amount of pidilizumab; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating pancreatic cancer, the method comprising: (a) administering to the subject a therapeutically effective amount of pidilizumab in combination with gemcitabine; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating renal cell carcinoma, the method comprising: (a) administering to the subject a therapeutically effective amount of pidilizumab in combination with dendritic cell/RCC fusion cell vaccine; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • Adoptive cell therapies are a useful approach for treating cancer.
  • Adoptive cell transfer refers to the passive transfer of ex vivo grown cells, often immune-derived cells, into a host with the aim of transferring the immunologic functionality and characteristics of the transplant.
  • Adoptive cell transfer can be autologous, as is common in adoptive T-cell therapies, or allogeneic.
  • TILs tumor infiltrating lymphocytes
  • TILs tumor infiltrating lymphocytes
  • peripheral blood mononuclear cells has been used to successfully treat patients with advanced solid tumors such as melanoma as well as patients with CD19-expressing hematologic malignancies.
  • Exemplary cell types for use in ACT include, without limitation, T-cells (e.g., CD8+ cells, CD4+ cells, etc.), NK-cells, delta-gamma T-cells, regulatory T-cells and peripheral blood mononuclear cells.
  • T-cells e.g., CD8+ cells, CD4+ cells, etc.
  • NK-cells e.g., NK-cells
  • delta-gamma T-cells e.g., regulatory T-cells and peripheral blood mononuclear cells.
  • TIL therapy TCR therapy
  • HLA human leukocyte antigen
  • Another way is the modification of cells with artificial molecules such as chimeric antigen receptors (CAR), commonly known as CAR-T cell therapy.
  • CAR chimeric antigen receptors
  • single chain antibodies can be used and CARs can also incorporate co-stimulatory domains.
  • aspects of the presently disclosed subject matter involve the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in combination immunotherapy together with adoptive cellular therapy, for example, to enhance adoptive cellular therapy for the treatment of cancer.
  • the presently disclosed subject matter involves the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in combination immunotherapy together with adoptive cellular therapy, and optionally in combination with a third immunotherapy, a fourth immunotherapy, and/or a fifth immunotherapy, such as tumor vaccines, A2aR blockade, and/or checkpoint blockade.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an adoptive cellular therapy.
  • the adoptive cellular therapy is selected from the group consisting of T-cells, CD8+ cells, CD4+ cells, NK-cells, delta-gamma T-cells, marrow infiltrating lymphocytes (MILs), regulatory T-cells, and peripheral blood mononuclear cells.
  • the adoptive cellular therapy comprises a tumor infiltrating lymphocyte (TIL).
  • TIL tumor infiltrating lymphocyte
  • the adoptive cellular therapy comprises a T-cell receptor modified lymphocyte.
  • the adoptive cellular therapy comprises a chimeric antigen receptor modified lymphocyte. In some embodiments, the adoptive cellular therapy comprises a chimeric antigen receptor T (CAR-T) cell. In some embodiments, the adoptive cellular therapy comprises marrow infiltrating lymphocytes (MILs). In some embodiments, the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is marrow-infiltrating lymphocytes (MILs).
  • MILs marrow infiltrating lymphocytes
  • Adenosine A2a receptor (A2aR) blockade has been reviewed and is reported to enhance tumor vaccines, checkpoint blockade and adoptive T cell therapy (see, e.g., Powell et al. 2015). Accordingly, aspects of the presently disclosed subject matter involves the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in combination immunotherapy together with adenosine A2a receptor (A2aR) blockade, for example, to enhance A2aR blockade for the treatment of cancer.
  • metabolic reprogramming agents e.g., DON, DON prodrugs, etc.
  • the presently disclosed subject matter involves the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in combination immunotherapy together with A2aR blockade, and optionally in combination with a third immunotherapy, a fourth immunotherapy, or a fifth immunotherapy, such as tumor vaccines, checkpoint blockade and/or adoptive cell therapy.
  • metabolic reprogramming agents e.g., DON, DON prodrugs, etc.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is an adenosine A2aR blockade.
  • the second immunotherapy is an adenosine A2aR blockade.
  • Exemplary A2aR inhibitors of use in the A2aR blockade as an immunotherapy include, without limitation, SCH58261, SYN115, ZM241365 and FSPTP.
  • the presently disclosed subject matter provides a method for treatment of a subject with a CD73-expressing tumor, the method comprising: (a) administering a therapeutically effective amount of SCH58261 to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • exemplary CD73-expressing tumors include, without limitation, breast tumors (e.g., breast adenocarcinoma, metastatic breast cancer) and melanoma (e.g., metastatic).
  • the CD73-expressing tumor is a metastatic tumor and administration of SCH58261 suppresses metastases in the CD73-expressing tumor.
  • the CD73-expressing tumor is melanoma and SCH58261 and the metabolic reprogramming agent are administered in combination with an anti-PD1 antibody to prolong survival and reduce the metastatic melanoma burden.
  • the CD73-expressing tumor is breast cancer and SCH58261 and the metabolic reprogramming agent are administered in combination with an anti-PD1 antibody to prolong survival and reduce the metastatic breast cancer burden.
  • the CD73-expressing tumor is a breast cancer tumor and SCH58261 and the metabolic reprogramming agent are administered in combination with a chemotherapeutic agent (e.g., doxorubicin) to increase the sensitivity of the breast cancer tumor to the chemotherapeutic agent.
  • a chemotherapeutic agent e.g., doxorubicin
  • the presently disclosed subject matter provides a method for treatment of a subject with a CD73-expressing tumor, the method comprising: (a) administering a therapeutically effective amount of SYN115 to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • an anti-PD-1 antibody is administered in combination with SYN115 and the metabolic reprogramming agent.
  • the presently disclosed subject matter provides a method for treatment of melanoma, the method comprising: (a) administering a therapeutically effective amount of ZM241365 to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • an anti-CTLA4 antibody is administered in combination with ZM241365 and the metabolic reprogramming agent.
  • the presently disclosed subject matter provides a method for treatment of bladder cancer, the method comprising: (a) administering a therapeutically effective amount of FSPTP to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • FSPTP is administered via intratumoral injection.
  • aspects of the presently disclosed subject matter involve the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.) in combination immunotherapy together with killer-cell immunoglobulin-like receptor (KIR) blockade, for example, to enhance KIR blockade for the treatment of cancer.
  • metabolic reprogramming agents e.g., DON, DON prodrugs, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III), below.
  • KIR killer-cell immunoglobulin-like receptor
  • the presently disclosed subject matter involves the use of metabolic reprogramming agents (e.g., DON, DON prodrugs, etc.) in combination immunotherapy together with KIR blockade, and optionally in combination with a third immunotherapy, a fourth immunotherapy, and/or a fifth immunotherapy, such as tumor vaccines, adoptive cell therapy, A2aR blockade, and/or checkpoint blockade.
  • metabolic reprogramming agents e.g., DON, DON prodrugs, etc.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a KIR blockade.
  • Exemplary KIR inhibitors of use in the KIR blockade as an immunotherapy include, without limitation, IPH2102/BMS-986015 (lirilumab).
  • the presently disclosed subject matter provides a method of treating acute myeloid leukemia, the method comprising: (a) administering a therapeutically effective amount of lirilumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating a solid tumor, the method comprising: (a) administering a therapeutically effective amount of lirilumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the solid tumor is a melanoma tumor, and lirilumab is administered in combination with nivolumab.
  • the solid tumor is a non-small cell lung cancer tumor, and lirilumab is administered in combination with nivolumab.
  • the solid tumor is a gastrointestinal tumor and lirilumab is administered in combination with nivolumab. In some embodiments, the solid tumor is a squamous cell carcinoma of the head and neck tumor and lirilumab is administered in combination with nivolumab. In some embodiments, the solid tumor is a hepatocellular carcinoma tumor and lirilumab is administered in combination with nivolumab.
  • the presently disclosed subject matter provides a method of treating a hematological tumor, the method comprising: (a) administering a therapeutically effective amount of lirilumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the hematological tumor is relapsed and/or refractory non-Hodgkin's lymphoma and lirilumab is administered in combination with nivolumab.
  • the hematological tumor is relapsed and/or refractory Hodgkin's lympohoma and lirilumab is administered in combination with nivolumab. In some embodiments, the hematological tumor is relapsed and/or refractory multiple myeloma and lirilumab is administered in combination with nivolumab. In some embodiments, the hematological tumor is relapsed and/or refractory chromic myelogenous leukemia and lirilumab is administered in combination with nivolumab.
  • the presently disclosed subject matter provides a method of treating relapsed and/or refractory multiple myeloma post autologous transplant, the method comprising: (a) administering a therapeutically effective amount of lirilumab to the subject optionally in combination with elotuzumab; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the presently disclosed subject matter provides a method of treating relapsed and/or refractory acute myeloid leukemia, the method comprising: (a) administering a therapeutically effective amount of lirilumab to the subject optionally in combination with 5-azacytidine; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • Vaccines stimulate the body's immune system to attack abnormal or malignant ells, such as cancer, resulting in a reduction of the those cells.
  • Cancer or tumor vaccines typically contain a tumor antigen in an immunogenic formulation that stimulates tumor antigen-specific helper cells, CTLs and/or B cells.
  • exemplary formulations of vaccines include, without limitations, dendritic cells (e.g., autologous dendritic cells pulsed with tumor cells or antigens), heterologous tumor cells transfected with immunostimulant agents, such as GM-CSF, recombinant virus, and/or peptides or proteins administered with adjuvants, such as CpG.
  • aspects of the presently disclosed subject matter involve combination immunotherapy using metabolic reprogramming agents (e.g., that decrease glutamine metabolism (e.g., DON or a DON prodrug, e.g, compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)) sequentially or simultaneously with vaccines, for example, for the treatment of cancer.
  • metabolic reprogramming agents e.g., that decrease glutamine metabolism (e.g., DON or a DON prodrug, e.g, compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)
  • the metabolic reprogramming agents can help delay or stop cancer cell growth, cause tumor shrinkage, prevent cancer from recurring, and/or eliminate cancer cells that have not been eradicated by other treatments.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a vaccine (e.g., tumor vaccine).
  • a vaccine e.g., tumor vaccine
  • Exemplary such vaccines include, without limitation, peptide vaccines, dendritic cell (DC) vaccines, EGFRvIII vaccines, mesothilin vaccine, G-VAX, listeria vaccines, and a dentritic cell/RCC fusion cell vaccine.
  • DC dendritic cell
  • EGFRvIII vaccines EGFRvIII vaccines
  • mesothilin vaccine G-VAX
  • listeria vaccines listeria vaccines
  • a dentritic cell/RCC fusion cell vaccine include, without limitation, peptide vaccines, dendritic cell (DC) vaccines, EGFRvIII vaccines, mesothilin vaccine, G-VAX, listeria vaccines, and a dentritic cell/RCC fusion cell vaccine.
  • the presently disclosed subject matter provides a method for the treatment or prevention of a human papillomavirus (HPV)-associated cancer in a subject in need thereof, the method comprising: (a) administering to the subject a therapeutically effective amount of a recombinant HPV vaccine; and (b) simultaneously or sequentially administering a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • HPV human papillomavirus
  • the subject has an oncogenic HPV-type 6 infection and the rHPV vaccine comprises a rHPV type 6 vaccine. In some embodiments, the subject has an oncogenic HPV-type 11 infection and the rHPV vaccine comprises a rHPV type 11 vaccine. In some embodiments, the subject has an oncogenic HPV-type 16 infection and the rHPV vaccine comprises a rHPV type 16 vaccine. In some embodiments, the subject has an oncogenic HPV-type 18 infection and the rHPV vaccine comprises a rHPV type 18 vaccine. In some embodiments, the subject is a female and the cancer is selected from the group consisting of cervical, vaginal and vulvar cancer.
  • the subject is a female between the 9 and 26 years old. In some embodiments, the subject is a female between 9 and 26 years old and the HPV-associated cancer is cervical cancer. In some embodiments, the subject is a female between 9 and 26 years old and the HPV-associated cancer is vaginal cancer. In some embodiments, the subject is a female between 9 and 26 years old and the HPV-associated cancer is vulvar cancer. In some embodiments, the subject is male or female and the HPV-associated cancer comprises anal cancer. In some embodiments, the subject is a male or female between 9 and 26 years old and the HPV-associated cancer is anal cancer. In some embodiments, the HPV vaccine comprises GARDASIL.
  • the subject is a female between 10 and 25 years old and the HPV-associated cancer is cervical cancer.
  • the HPV vaccine comprises CERVARIX.
  • the HPV vaccine is administered intramuscularly via injection as a suspension.
  • the presently disclosed subject matter provides a method for the treatment or prevention of a hepatitis B-associated cancer in a subject in need thereof, the method comprising: (a) administering to the subject a therapeutically effective amount of a hepatitis B vaccine; and (b) simultaneously or sequentially administering a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject has a hepatitis B virus infection and the cancer is liver cancer.
  • aspects of the presently disclosed subject matter involve combination immunotherapy using metabolic reprogramming agents (e.g., that decrease glutamine metabolism (e.g., DON or a DON prodrug, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)) sequentially or simultaneously with passive immunotherapy antibodies for the treatment of cancer.
  • metabolic reprogramming agents e.g., that decrease glutamine metabolism (e.g., DON or a DON prodrug, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of the second immunotherapy to the subject, wherein the second immunotherapy is a passive immunotherapy antibody.
  • passive immunotherapy antibody refers to monoclonal antibodies targeted to cancer cell-surface specific antigens to provide cancer immunity without actively stimulating a patient's immune system.
  • Exemplary passive immunotherapy antibodies of use herein include, without limitation, bevacizumab (e.g., AVASTIN), cetuximab (e.g., ERBITUX), rituximab (e.g., RITUXAN), trastuzumab (e.g., HERCEPTIN), alemtuzumab (e.g., CAMPATH), ibritumomab tiuxetan (e.g., ZEVALIN), panitumumab (e.g., VECTIBIX).
  • bevacizumab e.g., AVASTIN
  • cetuximab e.g., ERBITUX
  • rituximab e.g., RITUXAN
  • trastuzumab e.g., HERCEPTIN
  • alemtuzumab e.g., CAMPATH
  • the presently disclosed subject matter provides a method for treatment of a subject with metastatic colorectal cancer, the method comprising: (a) administering a therapeutically effective amount of bevacizumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • bevacizumab is administered as a first- or second-line treatment in combination with intravenous 5-fluorouracil-based chemotherapy.
  • bevacizumab is administered as a second-line treatment in combination with fluoropyrimidine-based (combined with irinotecan or oxaliplatin) chemotherapy after cancer progression following a first-line treatment that includes bevacizumab.
  • the presently disclosed subject matter provides a method for treatment of a subject with non-small cell lung cancer, the method comprising: (a) administering a therapeutically effective amount of bevacizumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the non-small cell lung cancer comprises advanced nonsquamous non-small cell lung cancer and bevacizumab is administered to subjects who have not received chemotherapy for their advanced disease in combination with carboplatin and paclitaxel.
  • the presently disclosed subject matter provides a method for treatment of a subject with platinum-resistant ovarian cancer, the method comprising: (a) administering a therapeutically effective amount of bevacizumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject has had no more than two prior chemotherapy treatments and bevacizumab is used to treat platinum-resistant recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer in the subject in combination with paclitaxel, pegylated liposomal doxorubicin or topotecan.
  • the presently disclosed subject matter provides a method for treatment of a subject with advanced cervical cancer, the method comprising: (a) administering a therapeutically effective amount of bevacizumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject has persistent, recurrent, or metastatic cervical cancer and bevacizumab is administered in combination with paclitaxel and cisplatin.
  • the subject has persistent, recurrent, or metastatic cervical cancer and bevacizumab is administered in combination with paclitaxel and topotecan.
  • the presently disclosed subject matter provides a method for treatment of a subject with metastatic renal cell carcinoma, the method comprising: (a) administering a therapeutically effective amount of bevacizumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject has metastatic kidney cancer and bevacizumab is administered in combination with interferon alfa.
  • the presently disclosed subject matter provides a method for treatment of a subject with recurrent glioblastoma, the method comprising: (a) administering a therapeutically effective amount of bevacizumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject with recurrent glioblastoma is an adult.
  • the presently disclosed subject matter provides a method for treatment of a subject with head and neck cancer, the method comprising: (a) administering a therapeutically effective amount of cetuximab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject has locally or regionally advanced squamous cell carcinoma of the head and neck and cetuximab is administered in combination with radiotherapy.
  • the subject has recurrent locoregional disease or metastatic squamous cell carcinoma of the head and neck and cetuximab is administered in combination with platinum-based therapy with 5-FU.
  • the subject has recurrent or metastatic squamous cell carcinoma of the head and neck and has failed to respond to prior platinum-based therapy.
  • the presently disclosed subject matter provides a method for treatment of a subject with metastatic colorectal cancer, the method comprising: (a) administering a therapeutically effective amount of cetuximab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the metastatic colorectal cancer comprises KRAS wild-type, epidermal growth factor rector (EGFR)-expressing, metastatic colorectal cancer, and cetuximab is administered in combination with FOLFIRI (irinotecan, 5-fluorouracil, and lucovorin).
  • the metastatic colorectal cancer comprises KRAS wild-type, epidermal growth factor rector (EGFR)-expressing, metastatic colorectal cancer, the subject is refractory to irinotecan-based chemotherapy, and cetuximab is administered in combination with irinotecan.
  • the metastatic colorectal cancer comprises KRAS wild-type, epidermal growth factor rector (EGFR)-expressing, metastatic colorectal cancer, and the subject has failed to respond to oxaliplatin and irinotecan-based chemotherapy.
  • the metastatic colorectal cancer comprises KRAS wild-type, epidermal growth factor rector (EGFR)-expressing, metastatic colorectal cancer, and the subject is intolerant to irinotecan.
  • the presently disclosed subject matter provides a method for treatment of a subject with non-Hodgkin's lymphoma, the method comprising: (a) administering a therapeutically effective amount of rituximab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • the subject has recurrent or refractory low-grade or follicular CD20-positive non-Hodgkin's lymphoma. In some embodiment, the subject has newly diagnosed CD20-positive non-Hodgkin's lymphoma. In some embodiments, the subject has low-grade or follicular CD20-positive non-Hodgkin's lymphoma and responded to initial treatment with CVP chemotherapy (cyclophosphamide, vincristine and prednisone).
  • CVP chemotherapy cyclophosphamide, vincristine and prednisone
  • the subject has CD20-positive diffuse large B-cell non-Hodgkin's lymphoma and the rituximab is administered in combination with CHOP chemotherapy (cyclophosphamide, doxorubicin hydrochloride, vincristine and prednisolone).
  • CHOP chemotherapy cyclophosphamide, doxorubicin hydrochloride, vincristine and prednisolone.
  • the subject has newly diagnosed or recurrent CD20-positive chronic lymphocytic leukemia and the rituximab is administered in combination with FC chemotherapy (fludarabine and cyclophosphamide).
  • the presently disclosed subject matter provides a method for treatment of a subject with early-stage breast cancer that is human epidermal growth factor receptor 2-positive (HER2+), the method comprising: (a) administering a therapeutically effective amount of alemtuzumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • HER2+ human epidermal growth factor receptor 2-positive
  • alemtuzumab is administered to the subject as part of a treatment course comprising doxorubicin, cyclophosphamide and either paclitaxel or docetaxel. In some embodiments, alemtuzumab is administered to the subject with docetaxel and carboplatin. In some embodiments, alemtuxumab is administered to the subject after treatment with anhtracylcine-based therapy. In some embodiments, the HER2+ breast cancer has spread into the subject's lymph nodes.
  • the HER2+ breast cancer has not spread into the subject's lymph nodes and the cancer is estrogen receptor/progesterone receptor (ER/PR)-negative or has one high risk feature selected from the group consisting of a tumor size >2 cm, age ⁇ 35 years, or tumor grade of 2 or 3.
  • ER/PR estrogen receptor/progesterone receptor
  • the presently disclosed subject matter provides a method for treatment of a subject with B-cell chronic lymphocytic leukemia (B-CLL), the method comprising: (a) administering a therapeutically effective amount of alemtuzumab to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • B-CLL B-cell chronic lymphocytic leukemia
  • the presently disclosed subject matter provides a method for treatment of a subject with low-grade or follicular B-cell non-Hodgkin's lymphoma (NHL) that has relapsed during or after treatment with other anticancer drugs or newly diagnosed follicular NHL following a response to initial anticancer therapy, the method comprising: (a) administering a therapeutically effective amount of ibritumomab tiuxetan to the subject; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • NDL non-Hodgkin's lymphoma
  • administering ibritumomab tiuxetan comprises intravenous injection comprising two infusions of rituximab (e.g., to reduce the number of B-cells in the subject's blood) and one injection of Yttrium-90 ibritumomab tiuxetan.
  • the presently disclosed subject matter provides a method for treatment of a subject with wild-type KRAS (exon 2 in codons 12 or 13) metastatic colorectal cancer, the method comprising: (a) administering a therapeutically effective amount of panitumumab to the subject as a first-line therapy in combination with folinic acid, fluorouracil and oxaliplatin; and (b) sequentially or simultaneously administering to the subject a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • wild-type KRAS exon 2 in codons 12 or 13 metastatic colorectal cancer
  • the presently disclosed subject matter provides a method for treatment of a subject with wild-type KRAS (exon 2 in codons 12 or 13) metastatic colorectal cancer, the method comprising: (a) administering a therapeutically effective amount of panitumumab following disease progression after prior treatment with fluoropyrimidine, oxalipltin, and irinotecan-containing chemotherapy; and (b) sequentially or simultaneously administering a therapeutically effective amount of a metabolic reprogramming agent that decreases glutamine metabolism.
  • aspects of the presently disclosed subject matter involve the use of metabolic reprogramming agents (e.g., DON, DON-prodrugs, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)) in combination immunotherapy together with a second, third, fourth, and/or fifth immunotherapy, for example, to enhance the immunotherapy for the treatment of cancer.
  • metabolic reprogramming agents e.g., DON, DON-prodrugs, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of a third immunotherapy to the subject, wherein the third immunotherapy is selected from the group consisting of checkpoint blockade, adoptive cell therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the third immunotherapy is selected from the group consisting of checkpoint blockade, adoptive cell therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of a third and/or fourth immunotherapy to the subject, wherein the third and/or fourth immunotherapy is selected from the group consisting of checkpoint blockade, adoptive cell therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the third and/or fourth immunotherapy is selected from the group consisting of checkpoint blockade, adoptive cell therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the method of treating cancer further includes simultaneously or sequentially administering a therapeutically effective amount of a third, fourth, and/or fifth immunotherapy to the subject, wherein the third, fourth, and/or fifth immunotherapy is selected from the group consisting of checkpoint blockade, adoptive cell therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the third, fourth, and/or fifth immunotherapy is selected from the group consisting of checkpoint blockade, adoptive cell therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • metabolic reprogramming agents e.g., DON or a DON prodrug, e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)
  • DON or a DON prodrug e.g., compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III)
  • compounds having any one of formula (I), formula (IIA), formula (IIB), or formula (III) as an adjuvant to cancer therapy, for example, for the treatment or prevention of cancer.
  • the method of treating cancer further includes simultaneously or sequentially administering to the subject a therapeutically effective amount of a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and combinations thereof.
  • a cancer therapy selected from the group consisting of: (i) chemotherapy; (ii) photodynamic therapy; (iii) proton therapy; (iv) radiotherapy; (v) surgery; and combinations thereof.
  • the first immunotherapy, and the second immunotherapy if administered is/are administered to the subject in the absence of chemotherapy. In some embodiments, the first immunotherapy, and the second immunotherapy if administered, is/are administered to the subject in the absence of photodynamic therapy. In some embodiments, the first immunotherapy, and the second immunotherapy if administered, is/are administered to the subject in the absence of proton therapy. In some embodiments, the first immunotherapy, and the second immunotherapy if administered, is/are administered to the subject in the absence of radiotherapy. In some embodiments, the first immunotherapy, and the second immunotherapy if administered, is/are administered to the subject in the absence of surgery.
  • Certain of the methods and compositions described herein relate to the metabolic reprogramming of cells using at least one metabolic reprogramming agent described herein to treat conditions, diseases, or disorders that involve metabolically reprogrammed cells whose activation, function, growth, proliferation, and/or survival depends on increased activity of at least one, at least two, or at least three metabolic pathways selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis.
  • aspects of the methods and compositions described herein relate to the use of least one metabolic reprogramming agent described herein to treat conditions, diseases, or disorders that involve aberrant and/or excessive amounts of at least one, at least two, or at least three metabolic pathways selected from the group consisting of aberrant and/or excessive glutamine metabolism, aberrant and/or excessive glycolysis, or aberrant and/or excessive fatty acid synthesis.
  • metabolic reprogramming agent generally refers to an agent that modulates the metabolic activity of at least one metabolic pathway in a cell, for example, to alter activation, function, growth, proliferation, and/or survival of the cell.
  • modulate broadly means to cause or facilitate a qualitative or quantitative change, alteration, or modification in a molecule, a process, pathway, or phenomenon of interest. Without limitation, such change may be an increase, decrease, a change in binding characteristics, or change in relative strength or activity of different components or branches of the process, pathway, or phenomenon.
  • modulator broadly refers to any molecule or agent that causes or facilitates a qualitative or quantitative change, alteration, or modification in a process, pathway, or phenomenon of interest.
  • modulator comprises both inhibitors and activators of a metabolic pathway or target.
  • modulator comprises both inhibitors and activators of expression and/or activity of a component involved glutamine metabolism, a component involved in glycolysis, and/or a component involved in fatty acid metabolism (e.g., fatty acid synthesis or fatty acid oxidation).
  • modulation of a metabolic pathway refers to modulation of activity of at least one component of the metabolic pathway.
  • modulator of the metabolic pathway can be, for example, a receptor ligand (e.g., a small molecule, an antibody, a siRNA), a ligand sequestrant (e.g., an antibody, a binding protein), a modulator of phosphorylation of a pathway component or a combination of such modulators.
  • One of skill in the art can easily test an agent to determine if it modulates a metabolic pathway by assessing, for example, phosphorylation status of a receptor or expression or synthesis of downstream proteins or enzymes controlled by the pathway in cultured cells and comparing the results to cells not treated with a modulator.
  • a modulator is determined to be a metabolic pathway modulator if the level of phosphorylation of the receptor or expression of or synthesis of downstream proteins or enzymes in a culture of cells is reduced by at least 20% compared to the level of phosphorylation of the receptor or expression or synthesis of downstream proteins or enzymes in cells that are cultured in the absence of the modulator; preferably the level of phosphorylation or expression or synthesis of downstream proteins or enzymes is altered by at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% in the presence of a metabolic pathway modulator.
  • “decrease”, “reduced”, “reduction”, “decrease” or “inhibit” are all used herein generally to mean a decrease by a statistically significant amount.
  • “reduced”, “reduction”, “decrease” or “inhibit” means a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90%, where the decrease is less than 100%.
  • the decrease includes a 100% decrease (e.g. absent level as compared to a reference sample), or any decrease between 10-100% as compared to a reference level.
  • the terms “increased”, “increase”, “enhance” or “activate” are all used herein to generally mean an increase by a statically significant amount; for the avoidance of any doubt, the terms “increased”, “increase”, “enhance” or “activate” means an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • compositions, and agents contemplated herein modulate an immune response.
  • an immune response e.g., inhibiting an immunosuppressive pathway, e.g., via checkpoint blockade, A2aR blockade, KIR blockade, etc.
  • the methods, compositions, and agents contemplated herein can decrease the immune response by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80, 90%, or as much as 100% as compared to a reference level (e.g., an objective measure of the immune response before employing the method, composition, and/or agent).
  • the methods, compositions, and agents contemplated herein can increase the immune response by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80, 90%, or as much as 100%, at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level (e.g., an objective measure of the immune response before employing the method, composition, and/or agent).
  • a reference level e.g., an objective measure of the immune response before employing the method, composition, and/or agent.
  • Certain methods, compositions, and agents contemplated herein modulate growth, proliferation, metastasis, and invasion of malignant cells (e.g., cancer cells).
  • the methods, compositions, and agents contemplated herein can decrease the growth, proliferation, metastasis, invasion, and/or survival of malignant cells (e.g., cancer cells) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80, 90%, or as much as 100% as compared to a reference level (e.g., an objective measure of the growth, proliferation, metastasis, invasion, and/or survival of malignant cells before employing the method, composition, and/or agent).
  • a reference level e.g., an objective measure of the growth, proliferation, metastasis, invasion, and/or survival of malignant cells before employing the method, composition, and/or agent.
  • statically significant refers to statistical significance and generally means a two standard deviation (2SD) below normal, or lower, concentration of the marker.
  • 2SD two standard deviation
  • concentration of the marker refers to statistical evidence that there is a difference. It is defined as the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true. The decision is often made using the p-value.
  • modulates”, “modulating”, and “modulation” are used interchangeably and refer to any one or a combination of a decrease in glutamine metabolism, a decrease in glycolysis, and a decrease in fatty acid synthesis.
  • modulates”, “modulating”, and “modulation” are used interchangeably and refer to any one or a combination of an increase in glutamine metabolism, an increase in glycolysis, and an increase in fatty acid synthesis.
  • modulates”, “modulating”, and “modulation” are used interchangeably and refer to any one or a combination of an increase in oxidative phosphorylation.
  • Glutamine (2-amino-4-carbamoylbutanoic acid), is used by the cell for both bioenergetic and biosynthetic needs.
  • Glutamine can be used as an amino acid for protein synthesis, as a carbon source, or as the primary nitrogen donor for multiple essential biosynthetic reactions in the cell. Once taken up by the cell, much of the glutamine is converted to glutamate by mitochondrial glutaminase. Both glutamine and glutamate contribute to anabolic metabolism; glutamine supplies nitrogen for nucleotide and hexosamine synthesis while glutamate is the nitrogen donor for the synthesis of many nonessential amino acids.
  • Glutamate can be used to support the production of NADPH or converted to the metabolic intermediates pyruvate and ⁇ -ketoglutarate.
  • glutamine metabolism or “glutamine metabolic activity” refers to the chemical reactions, enzymes, and pathways involving glutamine.
  • glutamine metabolic pathway is a biochemical pathway that involves glutamine.
  • a metabolic reprogramming agent can modulate any of the chemical reactions, enzymes and/or pathways involving glutamine.
  • at least one metabolic reprogramming agent can modulate chemical reactions, enzymes and/or pathways that do not directly involve glutamine, such as the conversion of pyruvate to acetyl CoA or the citric acid cycle, but indirectly affect any of the chemical reactions, enzymes and/or pathways involving glutamine.
  • Certain methods, compositions, and metabolic reprogramming agents contemplated herein decrease glutamine metabolism in cells.
  • the methods, compositions, and agents contemplated herein can decrease glutamine metabolism in cells by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80, 90%, or as much as 100% as compared to a reference level (e.g., an objective measure of the glutamine metabolic activity before employing the method, composition, and/or agent).
  • a reference level e.g., an objective measure of the glutamine metabolic activity before employing the method, composition, and/or agent.
  • At least one metabolic reprogramming agent is a glutamine antagonist (i.e., an agent that decrease glutamine metabolism).
  • glutamine antagonist refers to an agent that blocks or interferes with the synthesis or use of glutamine in a cell, and preferably in a cell that is part of a living organism.
  • glutamine antagonist interferes with the synthesis of glutamine, it is meant that the antagonist acts to reduce the amount or rate of glutamine synthesis to less than the amount or rate that would be experienced in the absence of the glutamine antagonist.
  • the glutamine antagonist interferes with the use of glutamine, it is meant that the antagonist acts to inhibit or block a metabolic pathway downstream of glutamine, that is, a pathway in which glutamine acts as a precursor of one or more non-glutamine compounds, or that the antagonist acts to deplete glutamine in a cell or an organism by reacting the glutamine to form a non-glutamine product, or by reversibly or irreversibly binding with glutamine to reduce its availability.
  • At least one metabolic reprogramming agent of the presently disclosed subject matter can be a glutamine analog that interferes with a glutamine metabolic pathway, an antagonist that inhibits the synthesis of glutamine, a glutamine depleting enzyme, a compound that reacts with glutamine under intracellular conditions to form a non-glutamine product, an antagonist that inhibits glutamine uptake by cells, an agent that inhibits glutamine oxidation, an agent that inhibits a glutamine transporter, an agent that inhibits glutaminolysis (a series of biochemical reactions by which glutamine is lysed to glutamate, aspartate, carbon dioxide, pyruvate, lactate, alanine and/or citrate), or a glutamine binding compound that reduces the biological availability of glutamine.
  • glutamine analog that interferes with a glutamine metabolic pathway
  • an antagonist that inhibits the synthesis of glutamine a glutamine depleting enzyme
  • a compound that reacts with glutamine under intracellular conditions to form a non-glutamine product an antagonist that inhibits glut
  • a compound that is a useful metabolic reprogramming agent may have two or more of these characteristics.
  • a compound that is a glutamine analog that interferes with a glutamine metabolic pathway might also act as an antagonist that inhibits the synthesis of glutamine.
  • At least one metabolic reprogramming agent can be an antagonist that inhibits the synthesis of glutamine.
  • compounds having this activity include inhibitors of glutamine synthase (EC 6.3.1.2), such as L-methionine-DL-sulfoximine, and phosphinothricin; inhibitors of glutamate synthase (EC 1.4.1.13); inhibitors of amidophosphoribosyltransferase (EC 2.4.2.14); and inhibitors of glutamate dehydrogenase; and mixtures of any two or more of these.
  • At least one metabolic reprogramming agent can be a glutamine depleting enzyme.
  • glutamine depleting enzymes include carbamoyl-phosphate synthase (EC 6.3.5.5), glutamine-pyruvate transaminase (EC 2.6.1.15), glutamine-tRNA ligase (EC 6.1.1.18), glutaminase (EC 3.5.1.2), D-glutaminase (EC 3.5.1.35), glutamine N-acyltransferase (EC2.3.1.68), glutaminase-asparaginase (in particular glutaminase-asparaginase of Pseudomonas 7a and Acinatobacter sp.), and mixtures of any two or more of these.
  • At least one metabolic reprogramming agent can be a compound that reacts with glutamine under intracellular conditions to form a non-glutamine product.
  • a compound having this property is phenylbutyrate (See Darmaun et al., Phenylbutyrate-induce glutamine depletion in humans: effect on leucine metabolism, pp. E801-E807, in Glutamine Depletion and Protein Catabolism, Am. Physiol. Soc. (1998)).
  • Another example of a glutamine antagonist having this characteristic is phenylacetate (See, U.S. Pat. No. 6,362,226).
  • At least one metabolic reprogramming agent can be an antagonist that inhibits glutamine uptake by cells.
  • compounds having this property include alpha-methylaminoisobutyric acid (inhibits GynT plasma membrane glutamine transporter; See, Varoqui et al., J. Biol. Chem., 275(6):4049-4054 (2000), wortmannin, and LY-294002 (inhibits hepatic glutamine transporter; See, Pawlik et al., Am. J. Physiol. Gastrointest. Liver Physiol., 278:G532-G541 (2000)).
  • At least one metabolic reprogramming agent can be a glutamine binding compound that reduces the biological availability of glutamine.
  • At least one metabolic reprogramming agent can be a glutamine analog that interferes with a glutamine metabolic pathway (e.g., decreases glutamine metabolism/metabolic activity).
  • glutamine analog that interferes with a glutamine metabolic pathway (e.g., decreases glutamine metabolism/metabolic activity).
  • compounds that can act in this manner include acivicin (L-(alpha S,5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), DON (6-diazo-5-oxo-L-norleucine), azaserine, azotomycin, chloroketone (L-2-amino-4-oxo-5-chloropentanoic acid), N 3 -(4-methoxyfumaroyl)-L-2,3-diaminopropanoic acid (FMDP) (inactivates glucosamine-6-phosphate synthase (EC 2.6.1.16), See, Zg6dka
  • At least one metabolic reprogramming agent is selected from the group consisting of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV).
  • At least one metabolic reprogramming agent is a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway (e.g., decreases glutamine metabolism/metabolic activity).
  • at least one metabolic reprogramming agent is a prodrug of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid), azaserine, 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV).
  • Suitable exemplary prodrugs of acivicin, azaserine, DON, and L-DONV can be found in “Prodrugs of Glutamine Analogs” (PCT/US2016/044767 (WO 2017/023774 A1), and herein incorporated by reference in its entirety).
  • Glycolysis is the metabolic pathway that converts glucose into pyruvate with the concurrent production of ATP. Pyruvate is a metabolic intermediate that can then enter the tricarboxylic acid (TCA) cycle within mitochondria to produce NADH and FADH 2 .
  • TCA tricarboxylic acid
  • the first step in glycolysis is the phosphorylation of glucose by hexokinase to form glucose 6-phosphate.
  • At least one metabolic reprogramming agent can modulate any of the chemical reactions and/or enzymes involved in glycolysis. In some embodiments, at least one metabolic reprogramming agent can modulate chemical reactions, enzymes and/or pathways that do not directly involve glycolysis, but indirectly affect any of the chemical reactions, enzymes and/or pathways involving glycolysis. Certain methods, compositions, and metabolic reprogramming agents contemplated herein decrease glycolysis in cells.
  • the methods, compositions, and agents contemplated herein can decrease glycolysis in cells by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80, 90%, or as much as 100% as compared to a reference level (e.g., an objective measure of the glycolytic metabolic activity before employing the method, composition, and/or agent).
  • a reference level e.g., an objective measure of the glycolytic metabolic activity before employing the method, composition, and/or agent.
  • the term “glycolytic metabolic activity” refers to the chemical reactions and enzymes involving the glycolysis pathway.
  • At least one metabolic reprogramming agent of the presently disclosed subject matter can be an agent that interferes with glycolysis or a related pathway that affects glycolysis; an agent that inhibits the synthesis of pyruvate and/or one of the intermediate products of glycolysis; an agent that inhibits one or more of the enzymes involved in glycolysis, such as hexokinase, phosphoglucose isomerase, phosphofructokinase, fructose-bisphosphate aldolase, triophosphate isomerase, glyceraldehyde phosphate dehydrogenase, phosphoglycerate kinase, phosphoglycerate mutase, enolase, and/or pyruvate kinase; an agent that depletes glucose-6-phosphate, one of the rate-limiting products in glycolysis; an agent that inhibits glucose uptake and/or transport across the plasma membrane by cells; or a glucose binding compound that reduces the biological availability of glucose.
  • At least one metabolic reprogramming agent interferes or inhibits the expression and/or activity of hexokinase.
  • inhibitors of hexokinase include, but are not limited to, 2-deoxyglucose (2-DG), 3-bromopyruvate (3-BrPA), lonidamine (LND), sodium fluoride, and potassium fluoride.
  • at least one metabolic reprogramming agent is 2-deoxy-D-glucose (2-DG).
  • Fatty acid synthesis is the process in the cell that creates fatty acids from acetyl-CoA and malonyl-CoA precursors.
  • Fatty acid oxidation is the process by which fatty acid molecules are broken down in the mitochondria to generate acetyl-CoA, which enters the citric acid cycle, and NADH and FADH 2 , which are used in the electron transport chain.
  • the enzyme AMP-activated protein kinase (AMPK) plays a role in cellular energy homeostasis and is a stimulator of fatty acid oxidation.
  • At least one metabolic reprogramming agent can modulate any of the chemical reactions and/or enzymes involved in fatty acid synthesis and/or fatty acid oxidation. In some embodiments, at least one metabolic reprogramming agent can modulate chemical reactions, enzymes and/or pathways that do not directly involve fatty acid synthesis and/or fatty acid oxidation, but indirectly affect any of the chemical reactions, enzymes and/or pathways involving fatty acid synthesis and/or fatty acid oxidation.
  • compositions, and metabolic reprogramming agents contemplated herein decrease fatty acid synthesis and/or increase fatty acid oxidation in cells.
  • the methods, compositions, and agents contemplated herein can decrease fatty acid synthesis and/or increase fatty acid oxidation in cells by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80, 90%, or as much as 100% as compared to a reference level (e.g., an objective measure of the synthesis of fatty before employing the method, composition, and/or agent).
  • At least one metabolic reprogramming agent of the presently disclosed subject matter can be an agent that interferes with fatty acid synthesis and/or fatty acid oxidation or a related pathway that affects fatty acid synthesis and/or fatty acid oxidation; an agent that increases fatty acid oxidation; an agent that increases one or more of the products of fatty acid oxidation; an agent that increases the expression and/or activity of one or more of the enzymes involved in fatty acid oxidation, such as acyl CoA dehydrogenase, enoyl CoA hydratase, 3-hydroxyacyl-CoA dehydrogenase, and ⁇ -ketothiolase; an agent that increases expression and/or activity of AMP-activated protein kinase (AMPK); an agent that increases uptake and/or transfer of activated fatty acids across the mitochondrial membrane; and an agent that increases the expression and/or activity of enzymes involved in the uptake and/or transfer of activated fatty acids across the mitochondrial membrane
  • At least one metabolic reprogramming agent is an activator of 5′ AMP-activated protein kinase (AMPK) activity.
  • AMPK 5′ AMP-activated protein kinase
  • At least one metabolic reprogramming agent that is an activator of AMPK activity can be an agent that increases concentrations of AMP in the cell; an AMP analogue, such as 5-amino-4-imidazolecarboxamide ribotide (ZMP); an agent that increases phosphorylation of AMPK, such as an agent that increases the expression and/or activity of a kinase that can phosphorylate AMPK; and an agent that is an allosteric modulator of AMPK, such as one that can modify AMPK to make it a better substrate for a kinase that can phosphorylate AMPK.
  • an AMP analogue such as 5-amino-4-imidazolecarboxamide ribotide (ZMP)
  • ZMP 5-amino-4-imidazolecarboxamide ribotide
  • an agent that increases phosphorylation of AMPK such as an agent that increases the expression and/or activity of a kinase that can phosphorylate
  • At least one metabolic reprogramming agent is metformin.
  • modulation of glutamine metabolism, glycolysis, and fatty acid metabolism may result in modulation of one or more genes or expression products of genes or biosynthesis or degradation of one or more enzymes.
  • expression means the process by which information from a gene or nucleic acid (e.g., DNA) is used in the synthesis of gene products (e.g., mRNA, RNA and/or proteins) and includes, but is not limited to, one or more of the steps of replication, transcription and translation.
  • the steps of expression which may be modulated by the agents contemplated herein may include, for example, transcription, splicing, translation and post-translational modification of a protein.
  • any particular protein may depend on the type of protein (e.g., protein kinase, transcriptional regulator, enzyme, etc.), its function (e.g., transcriptional regulation, catalysis, phosphorylation, signal transduction, etc.), and its subcellular localization (e.g., extracellular space, cytoplasm, nucleus, membrane, etc.).
  • protein kinase e.g., protein kinase, transcriptional regulator, enzyme, etc.
  • its function e.g., transcriptional regulation, catalysis, phosphorylation, signal transduction, etc.
  • subcellular localization e.g., extracellular space, cytoplasm, nucleus, membrane, etc.
  • appropriate agents to be used for modulation depending on the particular context (e.g., type of protein, biological function, subcellular localization, composition, method of use, mode of inhibition, etc.).
  • an agent can be used to inhibit enzymatic activity of an enzyme (e.g., at least one metabolic reprogramming agent that inhibits glutaminolysis catalyzed by glutaminase (e.g., a glutamine antagonist), at least one metabolic reprogramming agent that inhibits glycolysis catalyzed in part by hexokinase (e.g., 2-DG), etc.), inhibits the level or activity of phosphorylation of a protein kinase, inhibit activation of transcription or a signaling pathway.
  • an enzyme e.g., at least one metabolic reprogramming agent that inhibits glutaminolysis catalyzed by glutaminase (e.g., a glutamine antagonist), at least one metabolic reprogramming agent that inhibits glycolysis catalyzed in part by hexokinase (e.g., 2-DG), etc.
  • the metabolic reprogramming agents, chemotherapeutic agents, cytotoxic agents, immunotherapeutic agents, immunosuppressant agents, radiotherapeutic agents, anti-inflammatory agents, and neuroprotective agents described herein can be any type of agent.
  • agents that can be used as such agents in the methods, compositions, and uses described herein include small organic or inorganic molecules; saccharides; oligosaccharides; polysaccharides; a biological macromolecule selected from the group consisting of peptides, proteins, peptide analogs and derivatives; peptidomimetics; nucleic acids selected from the group consisting of siRNAs, shRNAs, antisense RNAs, ribozymes, dendrimers and aptamers; an extract made from biological materials selected from the group consisting of bacteria, plants, fungi, animal cells, and animal tissues; naturally occurring or synthetic compositions; microcarrier or nanocarrier consisting of one or more polymers, proteins, nucleic acids, lips, or metals; and any combination thereof.
  • small molecule can refer to agents that are “natural product-like,” however, the term “small molecule” is not limited to “natural product-like” agents. Rather, a small molecule is typically characterized in that it contains several carbon-carbon bonds, and has a molecular weight of less than 5000 Daltons (5 kD), preferably less than 3 kD, still more preferably less than 2 kD, and most preferably less than 1 kD. In some cases it is preferred that a small molecule have a molecular weight equal to or less than 700 Daltons.
  • RNA interference molecule refers to an agent which interferes with or inhibits expression of a target gene or genomic sequence by RNA interference (RNAi).
  • RNA interfering agents include, but are not limited to, nucleic acid molecules including RNA molecules which are homologous to the target gene or genomic sequence, or a fragment thereof, short interfering RNA (siRNA), short hairpin or small hairpin RNA (shRNA), microRNA (miRNA) and small molecules which interfere with or inhibit expression of a target gene by RNA interference (RNAi).
  • polynucleotide is used herein interchangeably with “nucleic acid” to indicate a polymer of nucleosides.
  • a polynucleotide is composed of nucleosides that are naturally found in DNA or RNA (e.g., adenosine, thymidine, guanosine, cytidine, uridine, deoxyadenosine, deoxythymidine,
  • deoxyguanosine, and deoxycytidine joined by phosphodiester bonds.
  • the term encompasses molecules comprising nucleosides or nucleoside analogs containing chemically or biologically modified bases, modified backbones, etc., whether or not found in naturally occurring nucleic acids, and such molecules may be preferred for certain applications.
  • polynucleotide sequence as used herein can refer to the polynucleotide material itself and/or to the sequence information (e.g. the succession of letters used as abbreviations for bases) that biochemically characterizes a specific nucleic acid.
  • a polynucleotide sequence presented herein is presented in a 5′ to 3′ direction unless otherwise indicated.
  • the nucleic acid molecules that modulate the metabolic pathways or targets described herein can, in some embodiments, be inserted into vectors and used as gene therapy vectors.
  • Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Pat. No. 5,328,470) or by stereotactic injection (see e.g., Chen et al. Proc. Natl. Acad. Sci. USA 91:3054-3057, 1994).
  • the pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can include one or more cells which produce the gene delivery system.
  • polypeptide refers to a polymer of amino acids.
  • protein and “polypeptide” are used interchangeably herein.
  • a peptide is a relatively short polypeptide, typically between about 2 and 60 amino acids in length.
  • Polypeptides used herein typically contain amino acids, such as the 20 L-amino acids that are most commonly found in proteins. However, other amino acids and/or amino acid analogs known in the art can be used.
  • One or more of the amino acids in a polypeptide may be modified, for example, by the addition of a chemical entity, such as a carbohydrate group, a phosphate group, a fatty acid group, a linker for conjugation, functionalization, etc.
  • polypeptide that has a non-polypeptide moiety covalently or non-covalently associated therewith is still considered a “polypeptide”.
  • exemplary modifications include glycosylation and palmitoylation.
  • Polypeptides may be purified from natural sources, produced using recombinant DNA technology, synthesized through chemical means, such as conventional solid phase peptide synthesis, etc.
  • the term “polypeptide sequence” or “amino acid sequence” as used herein can refer to the polypeptide material itself and/or to the sequence information (e.g., the succession of letters or three letter codes used as abbreviations for amino acid names) that biochemically characterizes a polypeptide.
  • a polypeptide sequence presented herein is presented in an N-terminal to C-terminal direction unless otherwise indicated.
  • identity refers to the extent to which the sequence of two or more nucleic acids or polypeptides is the same.
  • the percent identity between a sequence of interest and a second sequence over a window of evaluation may be computed by aligning the sequences, determining the number of residues (nucleotides or amino acids) within the window of evaluation that are opposite an identical residue allowing the introduction of gaps to maximize identity, dividing by the total number of residues of the sequence of interest or the second sequence (whichever is greater) that fall within the window, and multiplying by 100.
  • fractions are to be rounded to the nearest whole number.
  • Percent identity can be calculated with the use of a variety of computer programs known in the art. For example, computer programs, such as BLAST2, BLASTN, BLASTP, Gapped BLAST, etc., generate alignments and provide percent identity between sequences of interest.
  • computer programs such as BLAST2, BLASTN, BLASTP, Gapped BLAST, etc.
  • the algorithm of Karlin and Altschul Karlin and Altschul, Proc. Natl. Acad. Sci. USA 87:22264-2268, 1990
  • Karlin and Altschul Proc. Natl. Acad. Sci. USA 90:5873-5877, 1993 is incorporated into the NBLAST and XBLAST programs of Altschul et al. (Altschul, et al., J. MoT Biol. 215:403-410, 1990).
  • Gapped BLAST is utilized as described in Altschul et al. (Altschul, et al. Nucleic Acids Res. 25: 3389-3402, 1997).
  • the default parameters of the respective programs may be used.
  • a PAM250 or BLOSUM62 matrix may be used.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (NCBI). See the Web site having URL www.ncbi.nlm.nih.gov for these programs.
  • percent identity is calculated using BLAST2 with default parameters as provided by the NCBI.
  • At least one metabolic reprogramming agent described herein can be used in combination with an additional therapeutic agent (e.g., a pharmaceutically active agent, e.g., a drug approved by a regulatory agency).
  • an additional therapeutic agent e.g., a pharmaceutically active agent, e.g., a drug approved by a regulatory agency.
  • the therapeutic agent may act synergistically with the agent described herein, or they may independently exert their intended effects.
  • the disclosure contemplates any therapeutic agent which a skilled artisan would use in connection with a method, use, or composition described herein.
  • therapeutic agents contemplated for use in the presently disclosed methods, uses and compositions in combination with the metabolic reprogramming agents include, but are not limited to, chemotherapeutic agents/chemotherapy, immunotherapeutic agents/immunotherapy, immunosuppressant agents, anti-inflammatory agents, neuroprotective agents, neuroregenerative agents, neurotrophic factors, radiotherapeutic agents/radiotherapy, proton therapy, photodynamic therapy, and stem and progenitor cells used to replace and/or repair endogenous populations of abnormal, harmful, or unhealthy cells.
  • Chemotherapy and chemotherapeutic agent are used synonymously herein.
  • a “chemotherapeutic agent” is used to connote a compound or composition that is administered in the treatment of cancer.
  • Chemotherapeutic agents contemplated for use in combination with at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents described herein include, but are not limited to, alkylating agents, such as thiotepa and cyclophosphamide; alkyl sulfonates, such as busulfan, improsulfan and piposulfan; aziridines, such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; nitrogen mustards, such as chlorambucil,
  • Chemotherapeutic agents also include anti-hormonal agents that act to regulate or inhibit hormone action on tumors, such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens, such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston)
  • the chemotherapeutic agent is a topoisomerase inhibitor.
  • Topoisomerase inhibitors are chemotherapy agents that interfere with the action of a topoisomerase enzyme (e.g., topoisomerase I or II).
  • Topoisomerase inhibitors include, but are not limited to, doxorubicin HCl, daunorubicin citrate, mitoxantrone HCl, actinomycin D, etoposide, topotecan HCl, teniposide, and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these.
  • the chemotherapeutic agent is an anti-metabolite.
  • An anti-metabolite is a chemical with a structure that is similar to a metabolite required for normal biochemical reactions, yet different enough to interfere with one or more normal functions of cells, such as cell division.
  • Anti-metabolites include, but are not limited to, gemcitabine, fluorouracil, capecitabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, 5-azacytidine, 6-mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these.
  • the chemotherapeutic agent is an antimitotic agent, including, but not limited to, agents that bind tubulin.
  • the agent is a taxane.
  • the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel.
  • the antimitotic agent comprises a vinca alkaloid, such as vincristine, binblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives thereof.
  • immunotherapeutic agent refers to a molecule that can aid in the treatment of a disease by inducing, enhancing, or suppressing an immune response in a cell, tissue, organ or subject.
  • immunotherapeutic agents contemplated for use in combination with at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents described herein include, but are not limited to, immune checkpoint molecules (e.g., antibodies to immune checkpoint proteins), interleukins (e.g., IL-2, IL-7, IL-12, IL-15), cytokines (e.g., interferons, G-CSF, imiquimod), chemokines (e.g., CCL3, CCL26, CXCL7), vaccines (e.g., peptide vaccines, dendritic cell (DC) vaccines, EGFRvIII vaccines, mesothilin vaccine, G-VAX, listeria vaccines), and adopt
  • immunosuppressant agent means an agent which may be used in immunotherapy to reduce or prevent an immune response in a cell, organ, tissue, or subject.
  • immunosuppressant agents contemplated for use in combination with at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents include, without limitation, corticosteriods, calcineurin inhibitors, antiproliferative agents, SIP receptor agonists, kinase inhibitors, monoclonal antilymphocyte antibodies and polyclonal antilymphocyte antibodies.
  • Non-limiting examples of corticosteroids include Prednisone (Deltasone® and Orasone®) and Methylprednisolone (SoluMedrol®).
  • Non-limiting examples of calcineurin inhibitors include Cyclosporine (Cyclosporin A, SangCya, Sandimmune®, Neoral®, Gengraf®), ISA, Tx247, ABT-281, ASM 981 and Tacrolimus (Prograf®, FK506).
  • Non-limiting examples of antiproliferative agents include Mycophenolate Mofetil (CellCept®), Azathioprene (Imuran®), and Sirolimus (Rapamune®).
  • Non-limiting examples of SIP receptor agonists include FTY 720 or analogues thereof.
  • Non-limiting examples of kinase inhibitors include mTOR kinase inhibitors, which are compounds, proteins or antibodies that target, decrease or inhibit the activity and/or function of members of the serine/threonine mTOR family.
  • rapamycin include, without limitation, CCI-779, ABT578, SAR543, rapamycin and derivatives or analogs thereof, including 40-O-(2-hydroxyethyl)-rapamycin, rapalogs, including AP23573, AP23464, AP23675 and AP23841 from Ariad, Everolimus (CERTICAN, RAD001), biolimus 7, biolimus 9 and sirolimus (RAPAMUNE).
  • Kinase inhibitors also include protein kinase C inhibitors, which include the compounds described the PCT publications WO 2005/097108 and WO 2005/068455, which are herein incorporated by reference in their entireties.
  • Non-limiting examples of monoclonal antilymphocyte antibodies include Muromonab-CD3 (Orthoclone OKT3®), Interleukin-2 Receptor Antagonist (Basiliximab, Simulect®), and Daclizumab (Zenapax®).
  • Non-limiting examples of polyclonal antilymphocyte antibodies include Antithymocyte globulin-equine (Atgam®) and Antithymocyte globulin-rabbit (RATG, Thymoglobulin®).
  • Other immunosuppressants include, without limitation, SERP-1, a serine protease inhibitor produced by malignant rabbit fibroma virus (MRV) and myxoma virus (MYX), described in US Patent Publication No. 2004/0029801, which is incorporated herein by reference.
  • Immunosuppressant agents can be classified according to their specific molecular mode of action.
  • the four main categories of immunosuppressant drugs currently used in treating patients with transplanted organs are the following.
  • Calcineurin inhibitors inhibit T-cell activation, thus preventing T-cells from attacking the transplanted organ.
  • Azathioprines disrupt the synthesis of DNA and RNA as well as the process of cell division.
  • Monoclonal antibodies inhibit the binding of interleukin-2, which in turn slows down the production of T-cells in the patient's immune system.
  • Corticosteroids suppress inflammation associated with transplant rejection.
  • Immunosuppressants can also be classified according to the specific organ that is transplanted.
  • Basiliximab (Simulect) is also used in combination with such other drugs as cyclosporine and corticosteroids in kidney transplants.
  • IL-2 blockers including Simulect from Novartis, FK506 or CyA, MMF, prednisone or Rapanmycin are also used in kidney transplants.
  • Daclizumab (Zenapax) is also used in combination with such other drugs as cyclosporin and corticosteroids in kidney transplants. Similar drugs are used in heart transplants, but anti-lymphocyte globulin (ALG) is often used instead of Simulect.
  • Muromonab CD3 (Orthoclone OKT3) is used along with cyclosporine in kidney, liver and heart transplants.
  • Tacrolimus (Prograf) is used in liver and kidney transplants. It is under study for bone marrow, heart, pancreas, pancreatic island cell and small bowel transplantation.
  • photodynamic therapy also known as photoradiation therapy, phototherapy, and photochemotherapy, refers to a treatment that uses photosensitizing agents in combination with light to kill cancer cells.
  • the photosensitizing agents kill cancer cells upon light activation.
  • proton therapy also known as proton beam therapy, refers to a treatment that uses a beam of protons to irradiate and kill cancer cells.
  • anti-inflammatory agent refers to an agent that may be used to prevent or reduce an inflammatory response or inflammation in a cell, tissue, organ, or subject.
  • exemplary anti-inflammatory agents contemplated for use in combination with at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents include, without limitation, steroidal anti-inflammatory agents, a nonsteroidal anti-inflammatory agent, or a combination thereof.
  • anti-inflammatory agents include clobetasol, alclofenac, alclometasone dipropionate, algestone acetonide, alpha amylase, amcinafal, amcinafide, amfenac sodium, amiprilose hydrochloride, anakinra, anirolac, anitrazafen, apazone, balsalazide disodium, bendazac, benoxaprofen, benzydamine hydrochloride, bromelains, broperamole, budesonide, carprofen, cicloprofen, cintazone, cliprofen, clobetasol propionate, clobetasone butyrate, clopirac, cloticasone propionate, cormethasone acetate, cortodoxone, deflazacort, desonide, desoximetasone, dexamethasone, dexamethasone acetate, dexamethasone di
  • neuroprotective agents include, without limitation, L-dopa, dopamine agonists (e.g., apomorphine, bromocriptine, pergolide, ropinirole, pramipexole, or cabergoline), adenosine A2a antagonists (Shah et al., Curr. Opin. Drug Discov. Devel.
  • dopamine agonists e.g., apomorphine, bromocriptine, pergolide, ropinirole, pramipexole, or cabergoline
  • adenosine A2a antagonists Shah et al., Curr. Opin. Drug Discov. Devel.
  • a “radiotherapeutic agent” refers to those agents conventionally adopted in the therapeutic field of cancer treatment and includes photons having enough energy for chemical bond ionization, such as, for instance, alpha (a), beta (1), and gamma ( ⁇ ) rays from radioactive nuclei as well as x-rays.
  • the radiation may be high-LET (linear energy transfer) or low-LET. LET is the energy transferred per unit length of the distance. High LET is said to be densely ionizing radiation and Low LET is said to be sparsely ionizing radiation. Representative examples of high-LET are neutrons and alpha particles. Representative examples of low-LET are x-ray and gamma rays.
  • Low LET radiation including both x-rays and ⁇ rays is most commonly used for radiotherapy of cancer patients.
  • the radiation may be used for external radiation therapy that is usually given on an outpatient basis or for internal radiation therapy that uses radiation that is placed very close to or inside the tumor.
  • the radiation source is usually sealed in a small holder called an implant. Implants may be in the form of thin wires, plastic tubes called catheters, ribbons, capsules, or seeds. The implant is put directly into the body. Internal radiation therapy may require a hospital stay.
  • the ionizing radiation source is provided as a unit dose of radiation and is preferably an x-ray tube since it provides many advantages, such as convenient adjustable dosing where the source may be easily turned on and off, minimal disposal problems, and the like.
  • the ionizing radiation source may also comprise a radioisotope, such as a solid radioisotopic source (e.g., wire, strip, pellet, seed, bead, or the like), or a liquid radioisotopic filled balloon.
  • a radioisotope such as a solid radioisotopic source (e.g., wire, strip, pellet, seed, bead, or the like), or a liquid radioisotopic filled balloon.
  • the balloon has been specially configured to prevent leakage of the radioisotopic material from the balloon into the body lumen or blood stream.
  • the ionizing radiation source may comprise a receptacle in the catheter body for receiving radioisotopic materials like pellets or liquids.
  • the radioisotopic material may be selected to emit ⁇ , ⁇ and ⁇ .
  • ⁇ and ⁇ radiations are preferred since they may be quickly absorbed by the surrounding tissue and will not penetrate substantially beyond the wall of the body lumen being treated. Accordingly, incidental irradiation of the heart and other organs adjacent to the treatment region can be substantially eliminated.
  • the total number of units provided will be an amount determined to be therapeutically effective by one skilled in treatment using ionizing radiation. This amount will vary with the subject and the type of malignancy or neoplasm being treated. The amount may vary but a patient may receive a dosage of about 30-75 Gy over several weeks.
  • Exemplary radiotherapeutic agents contemplated for use in combination with at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents include, factors that cause DNA damage, such as ⁇ -rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells.
  • factors that cause DNA damage such as ⁇ -rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells.
  • Other forms of DNA damaging factors are also contemplated, such as microwaves and UV-irradiation.
  • Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens.
  • the radiotherapeutic agent is selected from the group consisting of 47 Sc, 67 Cu, 90 Y, 109 Pd, 123 I, 125 I, 131 I, 186 Re, 188 Re, 199 Au 211 At, 212 Pb, 212 B, 32 P and 33 P, 71 Ge, 77 As, 103 Pb, 105 Rh, 111 Ag, 119 Sb, 121 Sn, 131 Cs, 143 Pr, 161 Tb, 177 Lu, 191 Os, 193 MPt, 197 H, 43 K, 43 K, 52 Fe, 57 Co, 67 Cu, 67 Ga, 68 Ga, 77 Br, 81 Rb/ 81 MKr, 87 MSr, 99 MTc, 111 In, 113 Mn, 27 Cs, 129 Cs,
  • an agent described herein can be administered with an antigen (e.g., to induce an immune response).
  • an adjuvant can be used in combination with the antigen.
  • An agent described herein can also be used in combination with an imaging agent.
  • An agent e.g., a metabolic reprogramming agent
  • An agent can be attached to imaging agents for imaging and diagnosis of various diseased organs, tissues or cell types.
  • the agent can be labeled or conjugated a fluorophore or radiotracer for use as an imaging agent.
  • imaging agents are known in the art, as are methods for their attachment to agents (e.g., attaching an imaging agent to a proteins or peptides using metal chelate complexes, radioisotopes, fluorescent markers, or enzymes whose presence can be detected using a colorimetric markers (such as, but not limited to, urease, alkaline phosphatase, (horseradish) hydrogen peroxidase and glucose oxidase)).
  • An agent may also be dual labeled with a radioisotope in order to combine imaging through nuclear approaches and be made into a unique cyclic structure and optimized for binding affinity and pharmacokinetics.
  • Such agents can be administered by any number of methods known to those of ordinary skill in the art including, but not limited to, oral administration, inhalation, subcutaneous (sub-q), intravenous (I.V.), intraperitoneal (LP.), intramuscular (I.M.), intrathecal injection, or intratumoral injection.
  • the methods, compositions, and uses described herein can be used alone or in combination with other techniques, to diagnose access and monitor and direct therapy of metabolic reprogramming disorders.
  • the imaging agent can be used for detecting and/or monitoring tumors or sites of metastasis in a subject.
  • an agent e.g., a metabolic reprogramming agent
  • Exemplary methods for detecting and/or monitoring an agent labeled with an imaging agent in vivo include Gamma Scintigraphy, Positron Emission Tomography (PET), Single Photon Emission Computer Tomography (SPECT), Magnetic Resonance Imaging (MRI), X-ray, Computer Assisted X-ray Tomography (CT), Near Infrared Spectroscopy, and Ultrasound. These techniques provide information regarding detection of neoplastic involvement, particularly of inaccessible nodes in subjects with malignant diseases. Knowledge on the size of the node and the filling of nodes can also be instructive.
  • PET Positron Emission Tomography
  • SPECT Single Photon Emission Computer Tomography
  • MRI Magnetic Resonance Imaging
  • X-ray X-ray
  • CT Computer Assisted X-ray Tomography
  • Ultrasound Ultrasound
  • agents or compositions targeted to the lymph nodes in detection applications will contain suitable contrast or imaging agents, such as ferromagnetic materials like iron oxide, perfluorochemicals such as perfluorooctylbromide, or gamma emitting radiolabels such as Technetium-99m, Indium-111, Gallium-67, Thallium-201, Iodine-131, 125, or 123, positron emitting radiolabels, such as Fluorine-18, or those produced by neutron activation, such as Samarium-153.
  • suitable contrast or imaging agents such as ferromagnetic materials like iron oxide, perfluorochemicals such as perfluorooctylbromide, or gamma emitting radiolabels such as Technetium-99m, Indium-111, Gallium-67, Thallium-201, Iodine-131, 125, or 123, positron emitting radiolabels, such as Fluorine-18, or those produced by neutron activation,
  • Imaging agents of use in the present disclosure include radioisotopes and dyes. Any conventional method according to radiolabeling which is suitable for labeling isotopes for in vivo use will be generally suitable for labeling detection agents according to the disclosure.
  • Internal detection procedures include intraoperative, intravascular or endoscopic, including laparoscopic, techniques, both surgically invasive and noninvasive. For example, when detecting a lymph node, a high signal-to-background ratio should to be achieved. Therapy also requires a high absolute accretion of the therapeutic agent in the lymph node, as well as a reasonably long duration of uptake and binding.
  • Suitable radioisotopes for the methods of the disclosure include: Actinium-225, Astatine-211, Iodine-123, Iodine-125, Iodine-126, Iodine-131, Iodine-133, Bismuth-212, Bromine-77, Indium-111, Indium-113m, Gallium-67, Gallium-68, Ruthenium-95, Ruthenium-97, Ruthenium-103, Ruthenium-105, Mercury-107, Mercury-203, Rhenium-186, Rhenium-188, Tellurium-121m, Tellurium-122m, Tellurium-125m, Thulium-165, Thulium-167, Thulium-168, Technetium-99m, Fluorine-18, Silver-111, Platinum-197, Palladium-109, Copper-67, Phosphorus-32, Phosphorus-33, Yttrium-90, Scandium-47, Samarium-153, Lutetium-177, Rhodium-105,
  • the most preferred radioisotope for use in the presently disclosed subject matter is Technetium-99m.
  • the radioisotope will emit a particle or ray in the 10-7,000 keV range, more preferably in the 50-1,500 keV range, and most preferably in the 80-250 keV range.
  • Isotopes preferred for external imaging include: Iodine-123, Iodine-131, Indium-111, Gallium-67, Ruthenium-97, Technetium-99m, Cobalt-57, Cobalt-58, Chromium-51, Iron-59, Selenium-75, Thallium-201, and Ytterbium-169.
  • Technetium-99m is the most preferred radioisotope for external imaging in the disclosure.
  • Isotopes most preferred for internal detection include: Iodine-125, Iodine-123, Iodine-131, Indium-111, Technetium-99m and Gallium-67. Technetium-99m is the most preferred isotope for internal detection.
  • the presently disclosed subject matter contemplates the use of at least one, at least two, or at least three metabolic reprogramming agents that decrease activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis, alone, or optionally together with one or more additional therapeutic agents described herein.
  • the presently disclosed subject matter involves the use of at least one metabolic reprogramming agent that decreases activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis for treating a condition, disease, or disorder that involves (i) metabolically reprogrammed cells whose activation, function, growth, proliferation, and/or survival depends on increased activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis or (ii) at least one of aberrant and/or excessive glutamine metabolism, aberrant and/or excessive glycolysis, or aberrant and/or excessive fatty acid synthesis.
  • the presently disclosed subject matter involves the use of at least two metabolic reprogramming agents. In some embodiments, the presently disclosed subject matter involves the use of at least three metabolic reprogramming agents.
  • the presently disclosed subject matter involves the use of at least one metabolic reprogramming agent that decreases glutamine metabolism as an immunotherapy to treat a cancer.
  • the presently disclosed subject matter involves the use of at least one metabolic reprogramming agent that decreases glutamine metabolism as an immunotherapy in combination with an additional immunotherapy to treat a cancer.
  • additional immunotherapy contemplated for use in combination with the at least one metabolic reprogramming agent include, without limitation, checkpoint blockade, adoptive cellular therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the presently disclosed subject matter involves the use of an effective amount of at least one metabolic reprogramming agent that decreases glutamine metabolism to treat lymphoma in a subject in need thereof.
  • the presently disclosed subject matter involves the use of an effective amount of at least one metabolic reprogramming agent that decreases glutamine metabolism to treat melanoma in a subject in need thereof.
  • a use described herein further comprises using an effective amount of at least one metabolic reprogramming agent that decreases glycolysis. In some embodiments, a use described herein further comprises uses an effective amount of at least one metabolic reprogramming agent that increases fatty acid oxidation.
  • compositions Comprising Metabolic Reprogramming Agents
  • compositions comprising one or more metabolic reprogramming agents for the treatment of certain conditions, diseases, and/or disorders involving metabolically reprogrammed cells.
  • the presently disclosed methods comprise the use of the presently disclosed metabolic reprogramming agents for the manufacture of a medicament for the treatment of certain conditions, diseases, and/or disorders involve metabolically reprogrammed cells.
  • the disclosure contemplates various pharmaceutical compositions comprising at least one, at least two, and or at least three metabolic reprogramming agents.
  • the presently disclosed subject matter provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one, at least two, or at least three metabolic reprogramming agents that decrease the activity of at least one metabolic pathway selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis, and a pharmaceutically acceptable carrier, diluent, or excipient.
  • the presently disclosed subject matter provides a pharmaceutical composition comprising at least one metabolic reprogramming agent that decreases glutamine metabolism as an immunotherapy to treat a cancer, and a pharmaceutically acceptable carrier, diluent, or excipient.
  • a pharmaceutical composition comprising at least one metabolic reprogramming agent, such as checkpoint blockade, adoptive cellular therapy, CAR-T cell therapy, marrow-infiltrating lymphocytes, A2aR blockade, KIR blockade, vaccines (e.g., tumor vaccines), passive immunotherapy antibodies, and combinations thereof.
  • the metabolic reprogramming composition comprises one or more additional therapeutic agents described herein.
  • the presently disclosed compositions can be administered to a subject for therapy by any suitable route of administration, including orally, nasally, transmucosally, ocularly, rectally, intravaginally, parenterally, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intra-articular, intra-sternal, intra-synovial, intra-hepatic, intralesional, intracranial, intraperitoneal, intranasal, intraocular injections, intratumoral injections, intracisternally, topically, as by powders, ointments or drops (including eyedrops), including buccally and sublingually, transdermally, through an inhalation spray, or other modes of delivery known in the art.
  • systemic administration means the administration of compositions comprising at least one metabolic reprogramming agent, such that it enters the patient's system and, thus, are subject to metabolism and other like processes, for example, subcutaneous administration.
  • parenteral administration and “administered parenterally” as used herein mean modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intarterial, intrathecal, intracapsular, intraorbital, intraocular, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection and infusion.
  • compositions can be manufactured in a manner known in the art, e.g. by means of conventional mixing, dissolving, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • the presently disclosed pharmaceutical compositions can be administered by rechargeable or biodegradable devices.
  • a variety of slow-release polymeric devices have been developed and tested in vivo for the controlled delivery of drugs, including proteinacious biopharmaceuticals.
  • Suitable examples of sustained release preparations include semipermeable polymer matrices in the form of shaped articles, e.g., films or microcapsules.
  • Sustained release matrices include polyesters, hydrogels, polylactides (U.S. Pat. No.
  • Sustained release compositions also include liposomally entrapped compositions comprising at least one metabolic reprogramming agent which can be prepared by methods known in the art (Epstein et al. (1985) Proc. Natl. Acad. Sci. U.S.A. 82:3688; Hwang et al. (1980) Proc. Natl. Acad. Sci. U.S.A. 77:4030; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324A).
  • the liposomes are of the small (about 200-800 angstroms) unilamelar type in which the lipid content is greater than about 30 mol % cholesterol, the selected proportion being adjusted for the optimal therapy.
  • Such materials can comprise an implant, for example, for sustained release of the presently disclosed compositions, which, in some embodiments, can be implanted at a particular, pre-determined target site.
  • the presently disclosed pharmaceutical compositions may comprise PEGylated therapeutics (e.g., PEGylated antibodies).
  • PEGylation is a well established and validated approach for the modification of a range of antibodies, proteins, and peptides and involves the attachment of polyethylene glycol (PEG) at specific sites of the antibodies, proteins, and peptides (Chapman (2002) Adv. Drug Deliv. Rev. 54:531-545).
  • PEGylation results include: (a) markedly improved circulating half-lives in vivo due to either evasion of renal clearance as a result of the polymer increasing the apparent size of the molecule to above the glomerular filtration limit, and/or through evasion of cellular clearance mechanisms; (b) improved pharmacokinetics; (c) improved solubility—PEG has been found to be soluble in many different solvents, ranging from water to many organic solvents, such as toluene, methylene chloride, ethanol and acetone; (d) PEGylated antibody fragments can be concentrated to 200 mg/ml, and the ability to do so opens up formulation and dosing options, such as subcutaneous administration of a high protein dose; this is in contrast to many other therapeutic antibodies which are typically administered intravenously; (e) enhanced proteolytic resistance of the conjugated protein (Cunningham-Rundles et.
  • compositions for parenteral administration include aqueous solutions of compositions comprising at least one metabolic reprogramming agent.
  • the presently disclosed pharmaceutical compositions can be formulated in aqueous solutions, for example, in some embodiments, in physiologically compatible buffers, such as Hank's solution, Ringer's solution, or physiologically buffered saline.
  • Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • suspensions of compositions include fatty oils, such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • the suspension also can contain suitable stabilizers or agents that increase the solubility of the compositions comprising at least one metabolic reprogramming agent to allow for the preparation of highly concentrated solutions.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art.
  • compositions for topical administration can be added to compositions for topical administration, as long as such ingredients are pharmaceutically acceptable and not deleterious to the epithelial cells or their function. Further, such additional ingredients should not adversely affect the epithelial penetration efficiency of the composition, and should not cause deterioration in the stability of the composition.
  • additional ingredients should not adversely affect the epithelial penetration efficiency of the composition, and should not cause deterioration in the stability of the composition.
  • fragrances, opacifiers, antioxidants, gelling agents, stabilizers, surfactants, emollients, coloring agents, preservatives, buffering agents, and the like can be present.
  • the pH of the presently disclosed topical composition can be adjusted to a physiologically acceptable range of from about 6.0 to about 9.0 by adding buffering agents thereto such that the composition is physiologically compatible with a subject's skin.
  • compositions are formulated into pharmaceutically acceptable dosage forms, such as described herein or by other conventional methods known to those of skill in the art.
  • the “effective amount” or “therapeutically effective amount” of an active agent or drug delivery device refers to the amount necessary to elicit the desired biological response.
  • the effective amount of an agent or device may vary depending on such factors as the desired biological endpoint, the agent to be delivered, the composition of the encapsulating matrix, the target tissue, and the like.
  • the term “combination” is used in its broadest sense and means that a subject is administered at least two agents. More particularly, the term “in combination” refers to the concomitant administration of two (or more) active agents for the treatment of a, e.g., single disease state.
  • the active agents may be combined and administered in a single dosage form, may be administered as separate dosage forms at the same time, or may be administered as separate dosage forms that are administered alternately or sequentially on the same or separate days.
  • the active agents are combined and administered in a single dosage form.
  • the active agents are administered in separate dosage forms (e.g., wherein it is desirable to vary the amount of one but not the other).
  • the single dosage form may include additional active agents for the treatment of the disease state.
  • compositions can be administered alone or in combination with adjuvants that enhance stability of the agents, facilitate administration of pharmaceutical compositions containing them in certain embodiments, provide increased dissolution or dispersion, increase activity, provide adjuvant therapy, and the like, including other active ingredients.
  • combination therapies utilize lower dosages of the conventional therapeutics, thus avoiding possible toxicity and adverse side effects incurred when those agents are used as monotherapies.
  • the timing of administration of at least one metabolic reprogramming agent can be varied so long as the beneficial effects of the combination of these agents are achieved. Accordingly, the phrase “in combination with” refers to the administration of at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents, and optionally additional agents either simultaneously, sequentially, or a combination thereof.
  • a subject administered a combination of at least one, at least two, or at least three metabolic reprogramming agents, and optionally additional agents can receive at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, and at least three metabolic reprogramming agents, and optionally additional agents at the same time (i.e., simultaneously) or at different times (i.e., sequentially, in either order, on the same day or on different days), so long as the effect of the combination of all agents is achieved in the subject.
  • agents administered sequentially can be administered within 1, 5, 10, 30, 60, 120, 180, 240 minutes or longer of one another. In other embodiments, agents administered sequentially, can be administered within 1, 2, 3, 4, 5, 10, 15, 20 or more days of one another. Where the agents are administered simultaneously, they can be administered to the subject as separate pharmaceutical compositions, each comprising either at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents, and optionally additional agents, or they can be administered to a subject as a single pharmaceutical composition comprising all agents.
  • the effective concentration of each of the agents to elicit a particular biological response may be less than the effective concentration of each agent when administered alone, thereby allowing a reduction in the dose of one or more of the agents relative to the dose that would be needed if the agent was administered as a single agent.
  • the effects of multiple agents may, but need not be, additive or synergistic.
  • the agents may be administered multiple times.
  • the two or more agents when administered in combination, can have a synergistic effect.
  • the terms “synergy,” “synergistic,” “synergistically” and derivations thereof, such as in a “synergistic effect” or a “synergistic combination” or a “synergistic composition” refer to circumstances under which the biological activity of a combination of an agent and at least one additional therapeutic agent is greater than the sum of the biological activities of the respective agents when administered individually.
  • Synergy can be expressed in terms of a “Synergy Index (SI),” which generally can be determined by the method described by F. C. Kull et al. Applied Microbiology 9, 538 (1961), from the ratio determined by:
  • SI Synergy Index
  • Q A is the concentration of a component A, acting alone, which produced an end point in relation to component A;
  • Q a is the concentration of component A, in a mixture, which produced an end point
  • Q B is the concentration of a component B, acting alone, which produced an end point in relation to component B;
  • Q b is the concentration of component B, in a mixture, which produced an end point.
  • a “synergistic combination” has an activity higher that what can be expected based on the observed activities of the individual components when used alone.
  • a “synergistically effective amount” of a component refers to the amount of the component necessary to elicit a synergistic effect in, for example, another therapeutic agent present in the composition.
  • the presently disclosed subject matter provides a pharmaceutical composition including at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, at least three metabolic reprogramming agents, and optionally additional agents, alone or in combination with one or more additional therapeutic agents in admixture with a pharmaceutically acceptable excipient.
  • the presently disclosed subject matter provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, at least three metabolic reprogramming agents, and optionally additional agents, and a pharmaceutically acceptable carrier.
  • the compounds of the disclosure can be formulated for a variety of modes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remington: The Science and Practice of Pharmacy (20 th ed.) Lippincott, Williams and Wilkins (2000).
  • compositions of the present disclosure in particular, those formulated as solutions, may be administered parenterally, such as by intravenous injection.
  • the compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration.
  • Such carriers enable the compounds of the disclosure to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject (e.g., patient) to be treated.
  • the agents of the disclosure also may be formulated by methods known to those of skill in the art, and may include, for example, but not limited to, examples of solubilizing, diluting, or dispersing substances, such as saline; preservatives, such as benzyl alcohol; absorption promoters; and fluorocarbons.
  • compositions suitable for use in the present disclosure include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Generally, the compounds according to the disclosure are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used. A non-limiting dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • the preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
  • instructing means providing directions for applicable therapy, medication, treatment, treatment regimens, and the like, by any means, but preferably in writing. Instructing can be in the form of prescribing a course of treatment, or can be in the form of package inserts or other written promotional material. Accordingly, aspects of the presently disclosed subject matter include instructing a patient to receive a method of treatment or use an agent to treat a metabolic reprogramming disorder described herein.
  • promoting means offering, advertising, selling, or describing a particular drug, combination of drugs, or treatment modality, by any means, including writing, such as in the form of package inserts. Promoting herein refers to promotion of a metabolic reprogramming agent for an indication, where such promoting is authorized by the Food and Drug Administration (FDA) as having been demonstrated to be associated with statistically significant therapeutic efficacy and acceptable safety in a population of subjects. In some embodiments, promoting is not authorized by the Food and Drug Administration (FDA) (or other health regulatory agency, such as the European Medicines Agency (EMA), and promoting is for an off-label use. Accordingly, aspects of the presently disclosed subject matter include promoting a method of treatment or use described herein.
  • FDA Food and Drug Administration
  • EMA European Medicines Agency
  • substituted refers to the ability, as appreciated by one skilled in this art, to change one functional group for another functional group on a molecule, provided that the valency of all atoms is maintained.
  • substituent may be either the same or different at every position.
  • the substituents also may be further substituted (e.g., an aryl group substituent may have another substituent off it, such as another aryl group, which is further substituted at one or more positions).
  • substituent groups or linking groups are specified by their conventional chemical formulae, written from left to right, they equally encompass the chemically identical substituents that would result from writing the structure from right to left, e.g., —CH 2 O— is equivalent to —OCH 2 —; —C( ⁇ O)O— is equivalent to —OC( ⁇ O)—; —OC( ⁇ O)NR— is equivalent to —NRC( ⁇ O)O—, and the like.
  • R groups such as groups R 1 , R 2 , and the like, or variables, such as “m” and “n”
  • R 1 and R 2 can be substituted alkyls, or R 1 can be hydrogen and R 2 can be a substituted alkyl, and the like.
  • a when used in reference to a group of substituents herein, mean at least one.
  • a compound is substituted with “an” alkyl or aryl, the compound is optionally substituted with at least one alkyl and/or at least one aryl.
  • the group may be referred to as “R-substituted.” Where a moiety is R-substituted, the moiety is substituted with at least one R substituent and each R substituent is optionally different.
  • R or group will generally have the structure that is recognized in the art as corresponding to a group having that name, unless specified otherwise herein.
  • certain representative “R” groups as set forth above are defined below.
  • a “substituent group,” as used herein, includes a functional group selected from one or more of the following moieties, which are defined herein:
  • hydrocarbon refers to any chemical group comprising hydrogen and carbon.
  • the hydrocarbon may be substituted or unsubstituted. As would be known to one skilled in this art, all valencies must be satisfied in making any substitutions.
  • the hydrocarbon may be unsaturated, saturated, branched, unbranched, cyclic, polycyclic, or heterocyclic.
  • Illustrative hydrocarbons are further defined herein below and include, for example, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, allyl, vinyl, n-butyl, tert-butyl, ethynyl, cyclohexyl, and the like.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight (i.e., unbranched) or branched chain, acyclic or cyclic hydrocarbon group, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent groups, having the number of carbon atoms designated (i.e., C 1 -C 10 means one to ten carbons, including 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10 carbons).
  • alkyl refers to C 1-20 inclusive, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20 carbons, linear (i.e., “straight-chain”), branched, or cyclic, saturated or at least partially and in some cases fully unsaturated (i.e., alkenyl and alkynyl) hydrocarbon radicals derived from a hydrocarbon moiety containing between one and twenty carbon atoms by removal of a single hydrogen atom.
  • saturated hydrocarbon groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, sec-pentyl, isopentyl, neopentyl, n-hexyl, sec-hexyl, n-heptyl, n-octyl, n-decyl, n-undecyl, dodecyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, and homologs and isomers thereof.
  • Branched refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain.
  • Lower alkyl refers to an alkyl group having 1 to about 8 carbon atoms (i.e., a C 1-8 alkyl), e.g., 1, 2, 3, 4, 5, 6, 7, or 8 carbon atoms.
  • Higher alkyl refers to an alkyl group having about 10 to about 20 carbon atoms, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • alkyl refers, in particular, to C 1-8 straight-chain alkyls. In other embodiments, “alkyl” refers, in particular, to C 1-8 branched-chain alkyls.
  • Alkyl groups can optionally be substituted (a “substituted alkyl”) with one or more alkyl group substituents, which can be the same or different.
  • alkyl group substituent includes but is not limited to alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl.
  • alkyl chain There can be optionally inserted along the alkyl chain one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is hydrogen, lower alkyl (also referred to herein as “alkylaminoalkyl”), or aryl.
  • substituted alkyl includes alkyl groups, as defined herein, in which one or more atoms or functional groups of the alkyl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon group, or combinations thereof, consisting of at least one carbon atoms and at least one heteroatom selected from the group consisting of O, N, P, Si and S, and wherein the nitrogen, phosphorus, and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N, P and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which alkyl group is attached to the remainder of the molecule.
  • Examples include, but are not limited to, —CH 2 —CH 2 —O—CH 3 , —CH 2 —CH 2 —NH—CH 3 , —CH 2 —CH 2 —N(CH 3 )—CH 3 , —CH 2 —S—CH 2 —CH 3 , —CH 2 —CH 25 —S(O)—CH 3 , —CH 2 —CH 2 —S(O) 2 —CH 3 , —CH ⁇ CH—O—CH 3 , —Si(CH 3 ) 3 , —CH 2 —CH ⁇ N—OCH 3 , —CH ⁇ CH—N(CH 3 )—CH 3 , O—CH 3 , —O—CH 2 —CH 3 , and —CN.
  • Up to two or three heteroatoms may be consecutive, such as, for example, —CH 2 —NH—OCH 3 and —CH 2 —O—Si(CH 3 ) 3 .
  • heteroalkyl groups include those groups that are attached to the remainder of the molecule through a heteroatom, such as —C(O)NR′, —NR′R′′, —OR′, —SR, —S(O)R, and/or —S(O 2 )R′.
  • heteroalkyl is recited, followed by recitations of specific heteroalkyl groups, such as —NR′R or the like, it will be understood that the terms heteroalkyl and —NR′R′′ are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted herein as excluding specific heteroalkyl groups, such as —NR′R′′ or the like.
  • Cyclic and “cycloalkyl” refer to a non-aromatic mono- or multicyclic ring system of about 3 to about 10 carbon atoms, e.g., 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms.
  • the cycloalkyl group can be optionally partially unsaturated.
  • the cycloalkyl group also can be optionally substituted with an alkyl group substituent as defined herein, oxo, and/or alkylene.
  • cyclic alkyl chain There can be optionally inserted along the cyclic alkyl chain one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is hydrogen, unsubstituted alkyl, substituted alkyl, aryl, or substituted aryl, thus providing a heterocyclic group.
  • Representative monocyclic cycloalkyl rings include cyclopentyl, cyclohexyl, and cycloheptyl.
  • Multicyclic cycloalkyl rings include adamantyl, octahydronaphthyl, decalin, camphor, camphane, and noradamantyl, and fused ring systems, such as dihydro- and tetrahydronaphthalene, and the like.
  • cycloalkylalkyl refers to a cycloalkyl group as defined hereinabove, which is attached to the parent molecular moiety through an alkyl group, also as defined above.
  • alkyl group also as defined above.
  • examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.
  • cycloheteroalkyl or “heterocycloalkyl” refer to a non-aromatic ring system, unsaturated or partially unsaturated ring system, such as a 3- to 10-member substituted or unsubstituted cycloalkyl ring system, including one or more heteroatoms, which can be the same or different, and are selected from the group consisting of nitrogen (N), oxygen (O), sulfur (S), phosphorus (P), and silicon (Si), and optionally can include one or more double bonds.
  • N nitrogen
  • O oxygen
  • S sulfur
  • P phosphorus
  • Si silicon
  • the cycloheteroalkyl ring can be optionally fused to or otherwise attached to other cycloheteroalkyl rings and/or non-aromatic hydrocarbon rings.
  • Heterocyclic rings include those having from one to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, in which the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • heterocylic refers to a non-aromatic 5-, 6-, or 7-membered ring or a polycyclic group wherein at least one ring atom is a heteroatom selected from O, S, and N (wherein the nitrogen and sulfur heteroatoms may be optionally oxidized), including, but not limited to, a bi- or tri-cyclic group, comprising fused six-membered rings having between one and three heteroatoms independently selected from the oxygen, sulfur, and nitrogen, wherein (i) each 5-membered ring has 0 to 2 double bonds, each 6-membered ring has 0 to 2 double bonds, and each 7-membered ring has 0 to 3 double bonds, (ii) the nitrogen and sulfur heteroatoms may be optionally oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above heterocyclic rings may be fused to an aryl or heteroaryl ring.
  • Representative cycloheteroalkyl ring systems include, but are not limited to pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidyl, piperazinyl, indolinyl, quinuclidinyl, morpholinyl, thiomorpholinyl, thiadiazinanyl, tetrahydrofuranyl, and the like.
  • cycloalkyl and “heterocycloalkyl”, by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl”, respectively. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocycloalkyl examples include, but are not limited to, 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.
  • cycloalkylene and “heterocycloalkylene” refer to the divalent derivatives of cycloalkyl and heterocycloalkyl, respectively.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • Alkyl groups which are limited to hydrocarbon groups are termed “homoalkyl.”
  • alkenyl refers to a monovalent group derived from a C 1-20 inclusive straight or branched hydrocarbon moiety having at least one carbon-carbon double bond by the removal of a single hydrogen molecule.
  • Alkenyl groups include, for example, ethenyl (i.e., vinyl), propenyl, butenyl, 1-methyl-2-buten-1-yl, pentenyl, hexenyl, octenyl, allenyl, and butadienyl.
  • cycloalkenyl refers to a cyclic hydrocarbon containing at least one carbon-carbon double bond.
  • Examples of cycloalkenyl groups include cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadiene, cyclohexenyl, 1,3-cyclohexadiene, cycloheptenyl, cycloheptatrienyl, and cyclooctenyl.
  • alkynyl refers to a monovalent group derived from a straight or branched C 1-20 hydrocarbon of a designed number of carbon atoms containing at least one carbon-carbon triple bond.
  • alkynyl include ethynyl, 2-propynyl (propargyl), 1-propynyl, pentynyl, hexynyl, and heptynyl groups, and the like.
  • alkylene by itself or a part of another substituent refers to a straight or branched bivalent aliphatic hydrocarbon group derived from an alkyl group having from 1 to about 20 carbon atoms, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • the alkylene group can be straight, branched or cyclic.
  • the alkylene group also can be optionally unsaturated and/or substituted with one or more “alkyl group substituents.” There can be optionally inserted along the alkylene group one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms (also referred to herein as “alkylaminoalkyl”), wherein the nitrogen substituent is alkyl as previously described.
  • alkylene groups include methylene (—CH 2 —); ethylene (—CH 2 —CH 2 —); propylene (—(CH 2 ) 3 —); cyclohexylene (—C 6 H 10 —); —CH ⁇ CH—CH ⁇ CH—; —CH ⁇ CH—CH 2 —; —CH 2 CH 2 CH 2 CH 2 —, —CH 2 CH ⁇ CHCH 2 —, —CH 2 CsCCH 2 —, —CH 2 CH 2 CH(CH 2 CH 2 CH 3 )CH 2 —, —(CH 2 ) q —N(R)—(CH 2 ) r —, wherein each of q and r is independently an integer from 0 to about 20, e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and R is hydrogen or lower alkyl; methylenedioxyl (—O—CH 2 —O—); and ethylenedioxyl (—O—(CH 2 —
  • An alkylene group can have about 2 to about 3 carbon atoms and can further have 6-20 carbons. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being some embodiments of the present disclosure.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
  • heteroalkylene by itself or as part of another substituent means a divalent group derived from heteroalkyl, as exemplified, but not limited by, —CH 2 —CH 2 —S—CH 2 —CH 2 — and —CH 2 —S—CH 2 —CH 2 —NH—CH 2 —.
  • heteroalkylene groups heteroatoms also can occupy either or both of the chain termini (e.g., alkyleneoxo, alkylenedioxo, alkyleneamino, alkylenediamino, and the like).
  • no orientation of the linking group is implied by the direction in which the formula of the linking group is written.
  • the formula —C(O)OR′— represents both —C(O)OR′— and —R′OC(O)—.
  • aryl means, unless otherwise stated, an aromatic hydrocarbon substituent that can be a single ring or multiple rings (such as from 1 to 3 rings), which are fused together or linked covalently.
  • heteroaryl refers to aryl groups (or rings) that contain from one to four heteroatoms (in each separate ring in the case of multiple rings) selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom.
  • Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinoly
  • arylene and heteroarylene refer to the divalent forms of aryl and heteroaryl, respectively.
  • aryl when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
  • arylalkyl and heteroarylalkyl are meant to include those groups in which an aryl or heteroaryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl, furylmethyl, and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like).
  • haloaryl as used herein is meant to cover only aryls substituted with one or more halogens.
  • heteroalkyl where a heteroalkyl, heterocycloalkyl, or heteroaryl includes a specific number of members (e.g. “3 to 7 membered”), the term “member” refers to a carbon or heteroatom.
  • a ring structure for example, but not limited to a 3-carbon, a 4-carbon, a 5-carbon, a 6-carbon, a 7-carbon, and the like, aliphatic and/or aromatic cyclic compound, including a saturated ring structure, a partially saturated ring structure, and an unsaturated ring structure, comprising a substituent R group, wherein the R group can be present or absent, and when present, one or more R groups can each be substituted on one or more available carbon atoms of the ring structure.
  • the presence or absence of the R group and number of R groups is determined by the value of the variable “n,” which is an integer generally having a value ranging from 0 to the number of carbon atoms on the ring available for substitution.
  • n is an integer generally having a value ranging from 0 to the number of carbon atoms on the ring available for substitution.
  • Each R group if more than one, is substituted on an available carbon of the ring structure rather than on another R group.
  • a dashed line representing a bond in a cyclic ring structure indicates that the bond can be either present or absent in the ring. That is, a dashed line representing a bond in a cyclic ring structure indicates that the ring structure is selected from the group consisting of a saturated ring structure, a partially saturated ring structure, and an unsaturated ring structure.
  • Substituents for alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl monovalent and divalent derivative groups can be one or more of a variety of groups selected from, but not limited to: —OR′, ⁇ O, ⁇ NR′, ⁇ N—OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —C(O)NR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′—C(O)NR′′R′′′, NR′′C(O)OR′,
  • R′, R′′, R′′′ and R′′′′ each may independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups.
  • an “alkoxy” group is an alkyl attached to the remainder of the molecule through a divalent oxygen.
  • each of the R groups is independently selected as are each R′, R′′, R′′′ and R′′′′ groups when more than one of these groups is present.
  • R′ and R′′ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring.
  • —NR′R′′ is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., —CF 3 and —CH 2 CF 3 ) and acyl (e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like).
  • haloalkyl e.g., —CF 3 and —CH 2 CF 3
  • acyl e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like.
  • exemplary substituents for aryl and heteroaryl groups are varied and are selected from, for example: halogen, —OR′, —NR′R′′, —SR′, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —C(O)NR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′—C(O)NR′′R′′′, —NR′′C(O)OR′, —NR—C(NR′R′′R′′′) ⁇ NR′′′′, —NR—C(NR′R′′) ⁇ NR′—S(O)R′, —S(O) 2 R′, —S(O) 2 NR′R′′, —NRSO 2 R′, —CN and —NO 2 , —R′, —OR′, —NR′R′′, —SR′, —SiR′R′′R
  • Two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally form a ring of the formula -T-C(O)—(CRR′) q —U—, wherein T and U are independently —NR—, —O—, —CRR′— or a single bond, and q is an integer of from 0 to 3.
  • two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ) r —B—, wherein A and B are independently —CRR′—, —O—, —NR—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 NR′— or a single bond, and r is an integer of from 1 to 4.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula —(CRR′) s —X′—(C′′R′′′) d —, where s and d are independently integers of from 0 to 3, and X′ is —O—, —NR′—, —S—, —S(O)—, —S(O) 2 —, or —S(O) 2 NR′—.
  • the substituents R, R′, R′′ and R′′′ may be independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • acyl refers to an organic acid group wherein the —OH of the carboxyl group has been replaced with another substituent and has the general formula RC( ⁇ O)—, wherein R is an alkyl, alkenyl, alkynyl, aryl, carbocylic, heterocyclic, or aromatic heterocyclic group as defined herein).
  • R is an alkyl, alkenyl, alkynyl, aryl, carbocylic, heterocyclic, or aromatic heterocyclic group as defined herein).
  • acyl specifically includes arylacyl groups, such as a 2-(furan-2-yl)acetyl)- and a 2-phenylacetyl group. Specific examples of acyl groups include acetyl and benzoyl.
  • Acyl groups also are intended to include amides, —RC( ⁇ O)NR′, esters, —RC( ⁇ O)OR′, ketones, —RC( ⁇ O)R′, and aldehydes, —RC( ⁇ O)H.
  • alkoxyl or “alkoxy” are used interchangeably herein and refer to a saturated (i.e., alkyl-O—) or unsaturated (i.e., alkenyl-O— and alkynyl-O—) group attached to the parent molecular moiety through an oxygen atom, wherein the terms “alkyl,” “alkenyl,” and “alkynyl” are as previously described and can include C 1-20 inclusive, linear, branched, or cyclic, saturated or unsaturated oxo-hydrocarbon chains, including, for example, methoxyl, ethoxyl, propoxyl, isopropoxyl, n-butoxyl, sec-butoxyl, tert-butoxyl, and n-pentoxyl, neopentoxyl, n-hexoxyl, and the like.
  • alkoxyalkyl refers to an alkyl-O-alkyl ether, for example, a methoxyethyl or an ethoxymethyl group.
  • Aryloxyl refers to an aryl-O— group wherein the aryl group is as previously described, including a substituted aryl.
  • aryloxyl as used herein can refer to phenyloxyl or hexyloxyl, and alkyl, substituted alkyl, halo, or alkoxyl substituted phenyloxyl or hexyloxyl.
  • Alkyl refers to an aryl-alkyl-group wherein aryl and alkyl are as previously described, and included substituted aryl and substituted alkyl.
  • exemplary aralkyl groups include benzyl, phenylethyl, and naphthylmethyl.
  • Alkyloxyl refers to an aralkyl-O— group wherein the aralkyl group is as previously described.
  • An exemplary aralkyloxyl group is benzyloxyl, i.e., C 6 H 5 —CH 2 —O—.
  • An aralkyloxyl group can optionally be substituted.
  • Alkoxycarbonyl refers to an alkyl-O—C( ⁇ O)— group.
  • exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, butyloxycarbonyl, and tert-butyloxycarbonyl.
  • Aryloxycarbonyl refers to an aryl-O—C( ⁇ O)— group.
  • exemplary aryloxycarbonyl groups include phenoxy- and naphthoxy-carbonyl.
  • Alkoxycarbonyl refers to an aralkyl-O—C( ⁇ O)— group.
  • An exemplary aralkoxycarbonyl group is benzyloxycarbonyl.
  • Carbamoyl refers to an amide group of the formula —C( ⁇ O)NH 2 .
  • Alkylcarbamoyl refers to a R′RN—C( ⁇ O)— group wherein one of R and R′ is hydrogen and the other of R and R′ is alkyl and/or substituted alkyl as previously described.
  • Dialkylcarbamoyl refers to a R′RN—C( ⁇ O)— group wherein each of R and R′ is independently alkyl and/or substituted alkyl as previously described.
  • carbonyldioxyl refers to a carbonate group of the formula —O—C( ⁇ O)—OR.
  • acyloxyl refers to an acyl-O— group wherein acyl is as previously described.
  • amino refers to the —NH 2 group and also refers to a nitrogen containing group as is known in the art derived from ammonia by the replacement of one or more hydrogen radicals by organic radicals.
  • acylamino and alkylamino refer to specific N-substituted organic radicals with acyl and alkyl substituent groups respectively.
  • aminoalkyl refers to an amino group covalently bound to an alkylene linker. More particularly, the terms alkylamino, dialkylamino, and trialkylamino as used herein refer to one, two, or three, respectively, alkyl groups, as previously defined, attached to the parent molecular moiety through a nitrogen atom.
  • alkylamino refers to a group having the structure —NHR′ wherein R′ is an alkyl group, as previously defined; whereas the term dialkylamino refers to a group having the structure —NR′R′′, wherein R′ and R′′ are each independently selected from the group consisting of alkyl groups.
  • trialkylamino refers to a group having the structure —NR′R′′R′′′, wherein R′, R′′, and R′′′ are each independently selected from the group consisting of alkyl groups. Additionally, R′, R′′, and/or R′′′ taken together may optionally be —(CH 2 ) k — where k is an integer from 2 to 6. Examples include, but are not limited to, methylamino, dimethylamino, ethylamino, diethylamino, diethylaminocarbonyl, methylethylamino, isopropylamino, piperidino, trimethylamino, and propylamino.
  • the amino group is —NR′R′′, wherein R′ and R′′ are typically selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • alkylthioether and thioalkoxyl refer to a saturated (i.e., alkyl-S—) or unsaturated (i.e., alkenyl-S— and alkynyl-S—) group attached to the parent molecular moiety through a sulfur atom.
  • thioalkoxyl moieties include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like.
  • “Acylamino” refers to an acyl-NH— group wherein acyl is as previously described.
  • “Aroylamino” refers to an aroyl-NH— group wherein aroyl is as previously described.
  • carbonyl refers to the —C( ⁇ O)— group, and can include an aldehyde group represented by the general formula R—C( ⁇ O)H.
  • carboxyl refers to the —COOH group. Such groups also are referred to herein as a “carboxylic acid” moiety.
  • halo refers to fluoro, chloro, bromo, and iodo groups. Additionally, terms, such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • hydroxyl refers to the —OH group.
  • hydroxyalkyl refers to an alkyl group substituted with an —OH group.
  • mercapto refers to the —SH group.
  • oxo as used herein means an oxygen atom that is double bonded to a carbon atom or to another element.
  • nitro refers to the —NO 2 group.
  • thio refers to a compound described previously herein wherein a carbon or oxygen atom is replaced by a sulfur atom.
  • thiohydroxyl or thiol refers to a group of the formula —SH.
  • sulfide refers to compound having a group of the formula —SR.
  • sulfone refers to compound having a sulfonyl group —S(O 2 )R.
  • sulfoxide refers to a compound having a sulfinyl group —S(O)R
  • ureido refers to a urea group of the formula —NH—CO—NH 2 .
  • Certain compounds of the present disclosure may possess asymmetric carbon atoms (optical or chiral centers) or double bonds; the enantiomers, racemates, diastereomers, tautomers, geometric isomers, stereoisometric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as D- or L- for amino acids, and individual isomers are encompassed within the scope of the present disclosure.
  • the compounds of the present disclosure do not include those which are known in art to be too unstable to synthesize and/or isolate.
  • the present disclosure is meant to include compounds in racemic, scalemic, and optically pure forms.
  • Optically active (R)- and (S)-, or D- and L-isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • the compounds described herein contain olefenic bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers.
  • structures depicted herein are also meant to include all stereochemical forms of the structure; i.e., the R and S configurations for each asymmetric center. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric mixtures of the present compounds are within the scope of the disclosure.
  • tautomer refers to one of two or more structural isomers which exist in equilibrium and which are readily converted from one isomeric form to another.
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures with the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C-enriched carbon are within the scope of this disclosure.
  • the compounds of the present disclosure may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure.
  • the compounds of the present disclosure may exist as salts.
  • the present disclosure includes such salts.
  • Examples of applicable salt forms include hydrochlorides, hydrobromides, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, tartrates (e.g. (+)-tartrates, ( ⁇ )-tartrates or mixtures thereof including racemic mixtures, succinates, benzoates and salts with amino acids, such as glutamic acid.
  • These salts may be prepared by methods known to those skilled in art.
  • base addition salts such as sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent or by ion exchange.
  • acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • salts of amino acids such as arginate and the like
  • salts of organic acids like glucuronic or galactunoric acids and the like.
  • Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • the neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents.
  • Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure.
  • the present disclosure provides compounds, which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present disclosure.
  • prodrugs can be converted to the compounds of the present disclosure by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present disclosure when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • the term “about,” when referring to a value can be meant to encompass variations of, in some embodiments, ⁇ 100% in some embodiments ⁇ 50%, in some embodiments ⁇ 20%, in some embodiments ⁇ 10%, in some embodiments ⁇ 5%, in some embodiments ⁇ 1%, in some embodiments ⁇ 0.5%, and in some embodiments ⁇ 0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods or employ the disclosed compositions.
  • FIG. 3 shows that DON conditioned B16 melanoma to be killed by immunotherapy by inhibiting tumor infiltrating regulatory T cells (Foxp3 + ).
  • the glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON, 1) has shown robust anti-cancer efficacy in preclinical and clinical studies, but its development was halted due to marked systemic toxicities.
  • DON inhibits glutamine metabolism and provides antitumor efficacy in a murine model of gliobastoma, although toxicity was observed.
  • prodrug strategy to increase its brain delivery and limit systemic exposure. While these dual moiety prodrugs exhibited rapid metabolism in mouse plasma, several provided excellent stability in monkey and human plasma.
  • the most stable compound (5c, methyl-POM_DON-isopropyl-ester) was evaluated in monkeys, where it achieved 10-fold enhanced brain:plasma ratio versus DON. This strategy may provide a path to DON utilization in GBM patients.
  • GBM Glioblastoma multiforme
  • TemodarTM DNA-alkylating agent
  • GliadelTM carmustine releasing polymer GliadelTM
  • GBMs and other cancers rapidly and persistently proliferate often outgrowing vascular supplies necessitates that they develop a specialized metabolism (Schulze, et al., 2012; Hensley, et al., 2013).
  • Glutamine metabolism plays an essential role in this specialized metabolism by entering the TCA cycle and then contributing to the biosynthetic pathways (nucleotide, protein and lipid synthesis) necessary for unchecked cell growth, (Hensley, et al., 2013) rendering cancer cells dependent on glutamine.
  • GLS1 glutaminase
  • DON Because of its reactive diazo group, DON has demonstrated the ability to alkylate several glutamine-utilizing enzymes such as glutaminase, (Thangavelu, et al., 2014) NAD synthase, (Barclay, et al., 1966) and CTP synthetase (Hofer, et al., 2001) and FGAR aminotransferase (Grayzel, et al., 1960) in the purine and pyrimidine biosynthetic pathways, respectively.
  • glutaminase glutaminase
  • NAD synthase (Barclay, et al., 1966) and CTP synthetase (Hofer, et al., 2001) and FGAR aminotransferase (Grayzel, et al., 1960) in the purine and pyrimidine biosynthetic pathways, respectively.
  • DON robustly inhibited the growth of glutamine-dependent human cancer cells in vitro, and reduced tumor size and improved survival rates in vivo (Ahluwalia, et al., 1990; Cervantes-Madrid, et al., 2015). Because of the robust preclinical data, DON was evaluated in several clinical studies where it demonstrated promising results (Eagan, et al., 1982; Earhart, et al., 1982; Lynch, et al., 1982; Magill, et al., 1957; Rahman, et al., 1985; Sklaroff, et al., 1980; Sullivan, et al., 1962).
  • DON administration caused >50% regression or stable disease in late stage adult patients (Magill, et al., 1957) and in children with hematological malignancies or solid tumors (Sullivan, et al., 1988).
  • GI-related Magill, et al., 1957; Rahman, et al., 1985; Earhart, et al., 1990
  • the GI system is highly dependent on glutamine utilization.
  • DON prodrugs were synthesized using three types of amine promoieties including (oxodioxolenyl)methyl carbamate esters, dipeptides, and pivaloyl-oxy-methyl (POM)-based esters.
  • the dual promoiety-containing prodrugs resulted in sufficient chemical stability permitting further evaluation in in vitro metabolic stability assays. While all of the prodrugs exhibited rapid metabolism in mouse plasma, some provided surprising plasma stability in monkeys and humans.
  • the most stable DON prodrug (5c, methyl-POM-DON-isopropyl-ester) achieved an unexpected 10-fold enhanced brain: plasma ratio versus DON in monkeys, thus providing a possible clinical path to DON utilization in GBM patients.
  • prodrug strategy is often employed to enhance tissue penetration and change the pharmacokinetic parameters of effective drugs. Indeed, prodrug strategies are common in drug development as 5-7% of the approved worldwide drugs are prodrugs.
  • prodrug strategies are common in drug development as 5-7% of the approved worldwide drugs are prodrugs.
  • Our initial prodrug strategy for DON involved masking the carboxylic acid with simple alkyl esters such as ethyl 2a and isopropyl 2b. The synthesis of these two derivatives was straightforward affording compounds 2a and 2b in good yield. It is surprising to us that these simple DON alkyl esters had not previously been reported in the chemical literature, given that DON chemistry and utility has been described by numerous groups for over 60 years.
  • Table 2 outlines the plasma stability of representative DON. All prodrugs were completely metabolized in mouse plasma within the 60 min incubation time. However in monkey and human plasma, the prodrugs 5b and 5c, with methyl-POM on the amine and ethyl or isopropyl ester on the carboxylate respectively, demonstrated moderate/high stability with 60-75% of the prodrug remaining in monkey plasma, and 80-90% remaining in human plasma within the 60 min incubation time. Given that compound 5c (also referred to as compound 14b, see Table 1) had the best stability profile in human plasma, it was selected for further evaluation in pharmacokinetic studies and compared to DON for its ability to penetrate the brain and liberate DON.
  • mice Female athymic mice (RH_Foxn1nu mice) mice between 25 and 30 g were obtained (Harlan Sprague Dawley Inc, Indianapolis, Ind.), and maintained on a 12 hour light-dark cycle with ad libitum access to food and water.
  • H_Foxn1nu mice mice between 25 and 30 g were obtained (Harlan Sprague Dawley Inc, Indianapolis, Ind.), and maintained on a 12 hour light-dark cycle with ad libitum access to food and water.
  • U87 human glioma cells were injected s.c. (5 ⁇ 10 6 cells in 100 ml of PBS) in four separate locations on the flanks of each mouse.
  • mice When tumors grew to a mean volume of around 200 mm 3 , mice were randomized into either vehicle (HEPES-buffered saline, i.p.) or DON (1; 0.8 mg/kg, i.p.). In one cohort, mice were administered a single dose of the appropriate solution two hours after which glutamine levels were quantified in the tumor as described previously. 49 In brief, tumors were harvested, snap frozen, and homogenized in liquid N 2 then subjected to metabolite extraction using methanol and DI water.
  • vehicle HPES-buffered saline, i.p.
  • DON 0.8 mg/kg
  • mice All pharmacokinetic studies in mice were conducted according to protocol (#MO13M113) approved by the Animal Care and Use Committee at Johns Hopkins University.
  • C57BL/6 mice between 25 and 30 g were obtained from Harlan, and maintained on a 12 hour light-dark cycle with ad libitum access to food and water.
  • DON 1; 0.8 mg/kg, p.o. in phosphate-buffered saline
  • prodrug 5c at 0.8 mg/kg equivalent DON (1), p.o. in phosphate-buffered saline with 5% EtOH and 5% Tween-80).
  • mice were sacrificed by pentobarbital injection at 10, 30 and 90 minutes post drug administration, and blood was collected via cardiac puncture and placed into iced EDTA coated BD microtainers. Blood samples were spun at 2,000 g for 15 minutes, and plasma was removed and stored at ⁇ 80° C. Brain tissues were harvested following blood collection and immediately snap frozen in liquid nitrogen and stored at ⁇ 80° C. until LC/MS analysis.
  • ketamine Prior to drug administration, macaques were sedated with ketamine given as an intramuscular injection prior to test article administration. Sedation was maintained through blood and cerebrospinal fluid (CSF) sample collections with ketamine at a starting rate of 15 mg/kg with additional doses of 20-30 mg during the first hour. At subsequent time points ketamine was given at 10-15 mg/kg. DON (50 mM HEPES buffered saline) and 5c (Diastereoisomer 1), (50 mM HEPES buffered saline containing 5% ethanol and 5% tween) were administered (1.6 mg/kg equivalent) to the animals at a dosing volume of 1 mL/kg intravenously.
  • CSF cerebrospinal fluid
  • CSF sample target of 50 ⁇ L was obtained by percutaneous puncture of the cisterna magna at 30 min post dose.
  • Blood samples (1 mL) were collected at 15, 30, 1, 2 4 and 6 h post dose by percutaneous puncture of a peripheral vein.
  • Samples were processed for plasma (centrifuged at a temperature of 4° C., at 3,000 ⁇ g, for 10 minutes). All samples were maintained chilled on ice throughout processing. Samples were collected in microcentrifuge tubes, flash frozen, and placed in a freezer set to maintain ⁇ 80° C. until LC/MS analysis.
  • DON was extracted from samples (50 mg) with 250 ⁇ L methanol containing Glutamate-d5 (10 ⁇ M ISTD) by vortexing in low retention tubes. Samples were centrifuged at 16,000 ⁇ g for 5 minutes to precipitate proteins. Supernatants (200 ⁇ L) were moved to new tube and dried at 45° C. under vacuum for 1 hour.
  • Calibration curves over the range of 0.005-17.1 g/mL in plasma and CSF for DON were constructed from the peak area ratio of the analyte to the internal standard using linear regression with a weighting factor of 1/(nominal concentration). Correlation coefficient of greater than 0.99 was obtained in all analytical runs.
  • the mean predicted relative standard deviation for back calculated concentrations of the standards and QC's for all analytes were within the range of 85 to 115%, except for the lowest concentration which was within the range of 80 to 120% with an overall accuracy and precision of 6.7% and 6.6% respectively.
  • FIG. 8 illustrates that 25 (5 day dosing starting on day 7) is superior to CB-839 (30 day dosing starting day 1) in a CT26 tumor model.
  • FIG. 9 illustrates that 25 (4 days starting on day 6) is superior to CB-839 (continuous twice daily dosing starting on day 1 prior to engraftment) in a CT26 tumor model. Mice received daily 25 (1.9 mg/kg) on days 6-9 as compared to BID glutaminase inhibitor on days 1-15.
  • FIG. 10 illustrates that 25 (daily days 7-22) is superior to CB-839 (continuous twice daily dosing days 1-29) in a 4T1 breast cancer model. Mice received daily 25 (1.0 mg ⁇ kg/d) for days 7-22 as compared to BID glutaminase inhibitor for days 1-29.
  • DON pro-drugs demonstrate efficacy in multiple tumor types, including efficacy in B & T cell lymphomas, colon cancers, breast cancer and melanoma.
  • Daily dosing provides effective monotherapy. Every other day dosing leads to minimal resistance.
  • FIG. 11 illustrates that 25 dosing of 1 mg/kg following by 0.3 mg/kg leads to a complete and durable response in the MC38 tumor.
  • FIG. 12 illustrates that 25 provides a robust response and improved overall survival in multiple tumor models, including CT26 Colon Cancer.
  • FIG. 13 illustrates that 25 provides a robust response and improved overall survival in multiple tumor models, including 4T1 Breast Cancer.
  • FIG. 14 illustrates that mice cured with 25 alone immunologically reject tumors upon re-challenge, demonstrating that monotherapy with certain DON prodrugs, such as 25 monotherapy, is immunotherapy.
  • FIG. 15 additionally illustrates that 25 is immunotherapy.
  • DON/DON Prodrugs Condition Tumors to Immunotherapy and Significantly Enhance the Response to Checkpoint Inhibitors, Adoptive Cell Transfer and A2aR Inhibition
  • DON/DON prodrugs robustly enhance the immune mediated response to therapy with anti-PD1, the response to adoptive cell transfer, and the response to adenosine A2a receptor blockade.
  • Immunotherapy fails patients with rapidly progressive disease due to the lag in response. 30-40% of patients treated with anti-PD1 therapy progress rapidly in the first few months, chemotherapy showed an early survival advantage in NSCLC likely due to it's quick effect on rapidly growing disease compared to immunotherapy, and conditioning and adjuvant therapies to immunotherapy must be able to control tumor growth to be effective in these patients.
  • FIG. 16 illustrates that glutamine inhibition (e.g., using DON) reduces the oxygen consumption and lactate production of tumor cells.
  • FIG. 17 illustrates that glutamine inhibition (e.g., using DON) also improved the CD8/Treg ratio in the tumor and reduces hypoxia in the TILs.
  • FIG. 18 illustrates that 25 conditions tumors to be eliminated by anti-PD1 therapy in the MC38 Model, and that 25 rescues anti-PD1 failures.
  • FIG. 19 illustrates that even in the more difficult CT26 model, 25 unexpectedly enhances the response to anti-PD1.
  • FIG. 20 illustrates that inhibiting glutamine metabolism unexpectedly potentiates the anti-tumor response to A2aR inhibition.
  • FIG. 21 illustrates that inhibiting glutamine metabolism unexpectedly enhances the efficacy of adoptive cellular therapy (ACT) in a B16-OVA model.
  • ACT adoptive cellular therapy
  • Compound 14b was dissolved in 50 mM HEPES buffered saline containing 5% ethanol and 5% tween on the date of administration.
  • ketamine Prior to drug administration, macaques were sedated with ketamine given as an intramuscular injection prior to test article administration. Sedation was maintained through blood and cerebrospinal fluid (CSF) sample collections with ketamine at a starting rate of 15 mg/kg with additional doses of 20-30 mg during the first hour. At subsequent time points ketamine was given at 10-15 mg/kg.
  • CSF cerebrospinal fluid
  • DON 50 mM HEPES buffered saline
  • compound 14b 50 mM HEPES buffered saline containing 5% ethanol and 5% tween
  • CSF sample target of 50 ⁇ L was obtained by percutaneous puncture of the cisterna magna at 30 min post dose.
  • Blood samples (1 mL) were collected at 15 min, 30 min, 1 h, 2 h, 4 h, and 6 h post dose by percutaneous puncture of a peripheral vein.
  • Samples were processed for plasma (centrifuged at a temperature of 4° C., at 3,000 g, for 10 minutes). All samples were maintained chilled on ice throughout processing. Samples were collected in microcentrifuge tubes, flash frozen, and placed in a freezer set to maintain ⁇ 80° C. until LC/MS analysis.
  • DON was extracted from samples (50 mg) with 250 ⁇ L methanol containing glutamate-d 5 (10 ⁇ M ISTD) by vortexing in low retention tubes. Samples were centrifuged at 16,000 g for 5 minutes to precipitate proteins. Supernatants (200 ⁇ L) were moved to new tubes and dried at 45° C. under vacuum for 1 hour. To each tube, 50 ⁇ L of 0.2 M sodium bicarbonate buffer (pH 9.0) and 100 ⁇ L of 10 mM dabsyl chloride in acetone was added. After vortexing, samples were incubated at 60° C. for 15 minutes to derivatize.
  • the mean predicted relative standard deviation for back calculated concentrations of the standards and QC's for all analytes were within the range of 85 to 115%, except for the lowest concentration which was within the range of 80 to 120% with an overall accuracy and precision of 6.7% and 6.6% respectively.
  • DON The pharmacokinetics of DON and compound 14b in monkeys were evaluated.
  • i.v. administration of DON 1.6 mg/kg
  • compound 14b 3.6 mg/kg; 1.6 mg/kg DON equivalent
  • DON plasma profiles FIG. 40A .
  • DON administration provided high plasma exposures with AUC0-t of 42.7 nmol*h/mL.
  • compound 14b administration delivered ⁇ 7 fold lower plasma exposure of DON with AUC0-t of 5.71 nmol*h/mL.
  • the opposite observation was seen in the CSF where enhanced DON levels were observed after compound 14b administration.
  • Compound 47 was dissolved in a sterile saline containing 5% ethanol and 5% Tween 80 on the date of administration.
  • Swine studies were conducted under a protocol approved by the Johns Hopkins Animal Care and Use Committee.
  • Adult, female Göttingen x Yucutan miniature swine (Massachusetts General Hospital, MA) were housed in Johns Hopkins University facilities accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International in compliance with the Animal Welfare Act, Animal Welfare Regulations, and the Public Health Service Policy on the Humane Care and Use of Laboratory Animals.
  • Animals were maintained on a 14-h light and 10-h dark schedule, provided ad libitum water and a commercial miniswine diet (Teklad, Madison, Wis.) with environmental enrichment (fruit/vegetables) twice daily.
  • Blood samples (1 mL) were taken from CVC at predose, 5, 15, 30, 45, and 60 min. Plasma was separated by low speed centrifugation at 3000 g for 10 min at 4° C.
  • CSF was obtained from the cisterna magna using a 3.5 in ⁇ 22 gauge spinal needle (Becton Dickinson Health Care, Franklin Lakes, N.J., USA) at 60 min post-dose. All samples were flash frozen upon harvest and stored at ⁇ 80 C until bioanalysis.
  • DON Quantitation of DON in plasma, CSF, and brain homogenate by LC-MS/MS was performed. Briefly, DON was extracted from plasma, CSF, and brain samples with methanol containing glutamate-d 5 (1 ⁇ M ISTD) by vortexing followed by centrifugation 16000 g for 5 min. Supernatants were aliquoted and dried at 45° C. for under vacuum for 1 h. Sodium bicarbonate buffer (0.2M, pH 9.0) and dabsyl chloride (10 mM) in acetone were added to each tube, mixed, and incubated for 15 min at 60° C. to derivatize.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Emergency Medicine (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Oncology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
US15/885,275 2015-07-31 2018-01-31 Methods for cancer and immunotherapy using prodrugs of glutamine analogs Abandoned US20180221395A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/885,275 US20180221395A1 (en) 2015-07-31 2018-01-31 Methods for cancer and immunotherapy using prodrugs of glutamine analogs
US16/262,476 US10842763B2 (en) 2015-07-31 2019-01-30 Methods for cancer and immunotherapy using prodrugs of glutamine analogs
US17/102,341 US11759444B2 (en) 2015-07-31 2020-11-23 Methods for cancer and immunotherapy using prodrugs of glutamine analogs

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562199566P 2015-07-31 2015-07-31
US201562199381P 2015-07-31 2015-07-31
PCT/US2016/044829 WO2017023793A2 (en) 2015-07-31 2016-07-29 Methods for cancer and immunotherapy using glutamine analogues, including don
US15/885,275 US20180221395A1 (en) 2015-07-31 2018-01-31 Methods for cancer and immunotherapy using prodrugs of glutamine analogs

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/044829 Continuation-In-Part WO2017023793A2 (en) 2015-07-31 2016-07-29 Methods for cancer and immunotherapy using glutamine analogues, including don

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/262,476 Continuation-In-Part US10842763B2 (en) 2015-07-31 2019-01-30 Methods for cancer and immunotherapy using prodrugs of glutamine analogs

Publications (1)

Publication Number Publication Date
US20180221395A1 true US20180221395A1 (en) 2018-08-09

Family

ID=57943618

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/885,275 Abandoned US20180221395A1 (en) 2015-07-31 2018-01-31 Methods for cancer and immunotherapy using prodrugs of glutamine analogs
US15/885,147 Active US10568868B2 (en) 2015-07-31 2018-01-31 Methods and compositions for treating metabolic reprogramming disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/885,147 Active US10568868B2 (en) 2015-07-31 2018-01-31 Methods and compositions for treating metabolic reprogramming disorders

Country Status (5)

Country Link
US (2) US20180221395A1 (zh)
EP (2) EP3328376A4 (zh)
CN (2) CN108135875B (zh)
CA (2) CA2994256A1 (zh)
WO (2) WO2017023787A1 (zh)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10336778B2 (en) 2015-07-31 2019-07-02 The Johns Hopkins University Prodrugs of glutamine analogs
US10568868B2 (en) 2015-07-31 2020-02-25 The Johns Hopkins University Methods and compositions for treating metabolic reprogramming disorders
WO2020150639A1 (en) * 2019-01-18 2020-07-23 Dracen Pharmaceuticals, Inc. Combinaton therapy with a don prodrug and an immune checkpoint inhibitor
WO2020167831A1 (en) * 2019-02-11 2020-08-20 Dracen Pharmaceuticals, Inc. Method of preparing a don prodrug from l-glutamic acid
US10842763B2 (en) 2015-07-31 2020-11-24 The Johns Hopkins University Methods for cancer and immunotherapy using prodrugs of glutamine analogs
CN112807297A (zh) * 2021-01-20 2021-05-18 深圳市福田区风湿病专科医院 6-重氮-5-氧代-l-正亮氨酸在制备防治银屑病药物中的应用
US11760723B2 (en) 2019-02-11 2023-09-19 Dracen Pharmaceuticals, Inc. Method of preparing a DON prodrug from L-pyroglutamic acid
WO2024097320A1 (en) * 2022-11-02 2024-05-10 The Johns Hopkins University Azotomycin prodrugs with preferential tumor delivery

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10441654B2 (en) 2014-01-24 2019-10-15 Children's Hospital Of Eastern Ontario Research Institute Inc. SMC combination therapy for the treatment of cancer
WO2017023791A1 (en) * 2015-07-31 2017-02-09 The Johns Hopkins University Glutamine antagonists for the treatment of cognitive deficits
SG11201802830QA (en) 2015-10-05 2018-05-30 Calithera Biosciences Inc Combination therapy with glutaminase inhibitors and immuno-oncology agents
JP7061134B2 (ja) * 2017-03-20 2022-04-27 ユニバーシティ オブ サザン カリフォルニア 薬剤負荷ナノ粒子が表面に結合したcar t細胞の養子移入およびその使用
US20200289473A1 (en) * 2017-09-25 2020-09-17 The J David Gladstone Institutes A Testamentary Trust Established Under The Will Of J David Gladston Methods of treating neurodegeneration
DK3692015T3 (da) * 2017-10-06 2022-11-21 Univ Johns Hopkins Hidtil ukendte glutaminantagonister og anvendelser deraf
EP3703822A4 (en) * 2017-11-03 2021-11-10 Calithera Biosciences, Inc. CONJOINT THERAPY WITH GLUTAMINASE INHIBITORS
EP3517117A1 (en) * 2018-01-30 2019-07-31 Medizinische Universität Wien Medicament for prevention or treatment of rhinovirus infection
CN112969712A (zh) * 2018-07-23 2021-06-15 圣拉斐尔医院有限公司 联合糖基化抑制剂与car细胞疗法用于治疗癌症
JP7188936B2 (ja) * 2018-08-21 2022-12-13 国立大学法人富山大学 抗炎症剤
CN109045046A (zh) * 2018-09-12 2018-12-21 重庆医科大学 6-重氮-5-氧代-l-正亮氨酸在制备预防或治疗肝癌药物中的应用
WO2020210843A2 (en) * 2019-04-12 2020-10-15 The Johns Hopkins University Tolerogenic artificial antigen-presenting cells
KR102071614B1 (ko) * 2019-04-30 2020-01-30 김민청 6-디아조-5-옥소-l-노르루신을 포함하는 염증성 피부 질환 치료용 약제학적 조성물
WO2022022612A1 (en) * 2020-07-29 2022-02-03 Jacobio Pharmaceuticals Co., Ltd. Novel glutamine analogs
WO2022078462A1 (en) * 2020-10-15 2022-04-21 Jacobio Pharmaceuticals Co., Ltd. Novel glutamine analogs
JP2023546100A (ja) * 2020-10-15 2023-11-01 ジャコバイオ ファーマスーティカルズ カンパニー リミテッド 新規グルタミン類似体
CN114621120B (zh) * 2020-12-10 2023-11-03 深圳先进技术研究院 一种don前药分子、前药激活化合物和前药激活体系
WO2023081832A1 (en) * 2021-11-04 2023-05-11 The Johns Hopkins University Methods for preventing and treating acute kidney injury
CN114306620A (zh) * 2021-12-07 2022-04-12 深圳先进技术研究院 基于代谢检查点的人血清白蛋白纳米药物及其制备方法和应用
CN114344470A (zh) * 2022-01-26 2022-04-15 苏州大学 一种矽肺治疗靶点及其应用
WO2023202652A1 (en) * 2022-04-21 2023-10-26 Jacobio Pharmaceuticals Co., Ltd. Pharmaceutical combination and use thereof
CN114805138B (zh) * 2022-05-12 2023-10-10 中国科学院长春应用化学研究所 6-重氮-5-氧代-l-正亮氨酸的前药、其制备方法及应用
PL441816A1 (pl) * 2022-07-22 2024-01-29 Uniwersytet Medyczny w Łodzi Kompozycja farmaceutyczna zawierająca inhibitor białka GGT1 oraz antagonistę receptora CysLTR1 oraz jej zastosowanie w leczeniu inwazyjnych stadiów nowotworu

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
EP0102324A3 (de) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipide und Tenside in wässriger Phase
US4503044A (en) * 1983-04-25 1985-03-05 American Cyanamid Company Antibiotic LL-D05139 β
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
US5328470A (en) 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
WO2001041707A2 (en) 1999-12-08 2001-06-14 Vanderbilt University Modulation of in vivo glutamine and glycine levels in the treatment of autism
CA2404431C (en) 2000-03-27 2011-06-07 Thomas Jefferson University Guanylyl cyclase c in the detection of stomach and esophageal cancers
CA2423313A1 (en) 2000-09-29 2002-04-04 Z. Robert Zhong Methods of treating inflammatory and immune reactions and compositions therefor
WO2003096979A2 (en) * 2002-05-14 2003-11-27 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for celiac sprue
GB0314262D0 (en) * 2003-06-19 2003-07-23 Univ Nottingham Trent Novel compounds and methods of using the same
KR101140559B1 (ko) 2003-08-20 2012-07-05 제노포트 인코포레이티드 아실옥시알킬 카르바메이트 프로드러그, 합성 및 사용 방법
US7579313B2 (en) * 2003-11-18 2009-08-25 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
PT1709030E (pt) 2004-01-19 2008-07-01 Novartis Ag Derivados da indolilmaleimida
US20070232658A1 (en) 2004-04-08 2007-10-04 Jurgen Wagner Protein Kinase C Inhibitors for the Treatment of Autoimmune Diseases and of Transplant Rejiction
US20060276438A1 (en) * 2004-10-04 2006-12-07 Natarajan Sethuraman Prevention and treatment of influenza with glutamine antagonist agents
KR101205064B1 (ko) 2005-04-26 2012-11-27 에자이 알앤드디 매니지먼트 가부시키가이샤 암 면역요법을 위한 조성물과 방법
JP2009536173A (ja) * 2006-05-10 2009-10-08 メディカル エンザイムズ アクチエンゲゼルシャフト グルタドン
WO2008033572A1 (en) 2006-09-15 2008-03-20 Xenoport, Inc. Acyloxyalkyl carbamate prodrugs, methods of synthesis and use
US8343477B2 (en) 2006-11-01 2013-01-01 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
US20080160024A1 (en) * 2006-12-07 2008-07-03 Board Of Trustees Of The University Of Arkansas Inhibition of cancer metastasis
WO2009029729A1 (en) 2007-08-31 2009-03-05 Genelabs Technologies, Inc. Amino tricyclic-nucleoside compounds, compositions, and methods of use
EP3138581B1 (en) * 2011-03-17 2019-01-02 The University of Birmingham Re-directed immunotherapy
KR101900478B1 (ko) * 2011-08-04 2018-09-20 한올바이오파마주식회사 암의 재발 또는 전이의 억제용 약제학적 조성물
WO2014138391A1 (en) * 2013-03-06 2014-09-12 The Johns Hopkins University Targeting glutamine metabolism in brain tumors
WO2014160071A1 (en) 2013-03-14 2014-10-02 St. Jude Children's Research Hospital Methods and compositions for the treatment of glutamine-addicted cancers
JP6046529B2 (ja) 2013-03-21 2016-12-14 矢崎総業株式会社 端子
US10966998B2 (en) 2013-09-05 2021-04-06 The Johns Hopkins University Cancer therapy via a combination of epigenetic modulation and immune modulation
CN105828834A (zh) 2013-11-05 2016-08-03 同源生物服务股份有限公司 检查点抑制剂和治疗剂的组合以治疗癌症
CN112920180A (zh) * 2014-01-06 2021-06-08 理森制药股份公司 谷氨酰胺酶抑制剂
EP3116872A4 (en) 2014-03-14 2017-08-30 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
WO2015192014A1 (en) 2014-06-13 2015-12-17 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors
US10864180B2 (en) 2015-04-23 2020-12-15 The Johns Hopkins University Combination of immunotherapy with local chemotherapy for the treatment of malignancies
EP3328376A4 (en) 2015-07-31 2019-03-13 The Johns Hopkins University METHODS AND COMPOSITIONS FOR THE TREATMENT OF METABOLIC REPROGRAMMING DISORDERS
WO2017023791A1 (en) 2015-07-31 2017-02-09 The Johns Hopkins University Glutamine antagonists for the treatment of cognitive deficits
CN112920092B (zh) 2015-07-31 2024-04-26 约翰霍普金斯大学 谷氨酰胺类似物的前药

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10954257B2 (en) 2015-07-31 2021-03-23 The Johns Hopkins University Prodrugs of glutamine analogs
US10336778B2 (en) 2015-07-31 2019-07-02 The Johns Hopkins University Prodrugs of glutamine analogs
US11926640B2 (en) 2015-07-31 2024-03-12 The Johns Hopkins University Prodrugs of glutamine analogs
US10738066B2 (en) 2015-07-31 2020-08-11 The Johns Hopkins University Prodrugs of glutamine analogs
US11759444B2 (en) 2015-07-31 2023-09-19 The Johns Hopkins University Methods for cancer and immunotherapy using prodrugs of glutamine analogs
US10842763B2 (en) 2015-07-31 2020-11-24 The Johns Hopkins University Methods for cancer and immunotherapy using prodrugs of glutamine analogs
US10568868B2 (en) 2015-07-31 2020-02-25 The Johns Hopkins University Methods and compositions for treating metabolic reprogramming disorders
CN113438947A (zh) * 2019-01-18 2021-09-24 羿尊生物医药股份有限公司 Don前药和免疫检查点抑制剂的联合治疗
EP3911310A4 (en) * 2019-01-18 2022-10-12 Dracen Pharmaceuticals, Inc. COMBINATION THERAPY WITH A DON-PRODRUG AND AN IMMUNE CHECKPOINT INHIBITOR
WO2020150639A1 (en) * 2019-01-18 2020-07-23 Dracen Pharmaceuticals, Inc. Combinaton therapy with a don prodrug and an immune checkpoint inhibitor
US11760723B2 (en) 2019-02-11 2023-09-19 Dracen Pharmaceuticals, Inc. Method of preparing a DON prodrug from L-pyroglutamic acid
WO2020167831A1 (en) * 2019-02-11 2020-08-20 Dracen Pharmaceuticals, Inc. Method of preparing a don prodrug from l-glutamic acid
CN112807297A (zh) * 2021-01-20 2021-05-18 深圳市福田区风湿病专科医院 6-重氮-5-氧代-l-正亮氨酸在制备防治银屑病药物中的应用
WO2024097320A1 (en) * 2022-11-02 2024-05-10 The Johns Hopkins University Azotomycin prodrugs with preferential tumor delivery

Also Published As

Publication number Publication date
EP3328376A1 (en) 2018-06-06
CA2994256A1 (en) 2017-02-09
CN108135875B (zh) 2021-12-31
CA2994165A1 (en) 2017-02-09
CN108135875A (zh) 2018-06-08
WO2017023793A2 (en) 2017-02-09
EP3328376A4 (en) 2019-03-13
US10568868B2 (en) 2020-02-25
EP3328375B1 (en) 2022-08-31
CN108348492B (zh) 2021-09-28
WO2017023787A1 (en) 2017-02-09
CN108348492A (zh) 2018-07-31
US20180221337A1 (en) 2018-08-09
EP3328375A2 (en) 2018-06-06
EP3328375A4 (en) 2019-06-19

Similar Documents

Publication Publication Date Title
US20180221395A1 (en) Methods for cancer and immunotherapy using prodrugs of glutamine analogs
US11759444B2 (en) Methods for cancer and immunotherapy using prodrugs of glutamine analogs
ES2409755T3 (es) Método para Tratar el Cáncer Utilizando un Agente Anticanceroso Combinado
EP2786756B1 (en) Combination therapy with a topoisomerase inhibitor
TWI620565B (zh) 治療及預防移植物抗宿主病之方法
JP2016519684A (ja) 準最適に投与された薬物療法の有効性を改善するための及び/又は副作用を低減するための方法および組成物
US20210276964A1 (en) Compositions and methods for treating cancers
CN103347511B (zh) 化学治疗剂的抗肿瘤活性的增强剂
MX2013005196A (es) Metodos para tratamiento del cancer.
KR20140079831A (ko) 췌장암 및/또는 담도암 치료약
ES2848706T3 (es) Terapia de combinación contra el cáncer usando un compuesto azabicíclico
BR112021007283A2 (pt) método de tratamento de um câncer ou câncer refratário
WO2019113072A1 (en) Immune-stimulating peptides and checkpoint inhibitors, and uses thereof for treating cancer
AU2006246958A1 (en) Triazine compounds and compositions thereof for the treatment of cancers
KR101458233B1 (ko) 백혈병을 치료하기 위한 신규한 치료 용도
CN109528731B (zh) 具有协同作用治疗多发性骨髓瘤的药物组合物及其应用
ES2930009T3 (es) Métodos para el cáncer e inmunoterapia con análogos de glutamina, comprendiendo deoxinivalenol DON incluido
KR20090021211A (ko) 약물 투여 방법
JP7414230B2 (ja) 抗血液悪性腫瘍薬
AU2016277929A1 (en) Combination therapy using belinostat and pralatrexate to treat lymphoma
CN103083666A (zh) 一种药物组合物及其制剂和用途

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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