Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR TREATING METABOLIC REPROGRAMMING DISORDERS
Document Type and Number:
WIPO Patent Application WO/2017/023787
Kind Code:
A1
Abstract:
The presently disclosed subject matter relates to metabolic reprogramming agents that decrease glutamine metabolism, glycolysis, and fatty acid synthesis, pharmaceutical compositions comprising at least one, at least two, or at least three metabolic reprogramming agents, and the use of those agents and compositions for treating metabolic reprogramming disorders, such as immune disorders (e.g., autoimmune diseases), inflammatory diseases, and transplant rejection, pathologies due to CNS inflammation due to infection and not involving infection, and neurodegenerative disorders.

Inventors:
SLUSHER BARBARA (US)
POWELL JONATHAN (US)
Application Number:
PCT/US2016/044810
Publication Date:
February 09, 2017
Filing Date:
July 29, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV JOHNS HOPKINS (US)
International Classes:
A61K31/198; A61K31/357; A61K31/404; A61K31/421; A61K31/66; A61K45/06
Domestic Patent References:
WO2013019058A22013-02-07
WO2014160071A12014-10-02
WO2004113363A22004-12-29
WO2005097108A12005-10-20
WO2005068455A12005-07-28
Foreign References:
US20080160024A12008-07-03
US20090169537A12009-07-02
US6362226B22002-03-26
US5328470A1994-07-12
US20040029801A12004-02-12
US8946168B22015-02-03
US3773919A1973-11-20
EP0058481A11982-08-25
EP0133988A21985-03-13
US4485045A1984-11-27
US4544545A1985-10-01
EP0102324A21984-03-07
Other References:
LEVESLEY, J. ET AL.: "ABT-263 enhances sensitivity to metformin and 2-deoxyglucose in pediatric glioma by promoting apoptotic cell death", PLOS ONE, vol. 8, no. Issue 5, 2013, pages 1 - 12, XP055362775
VANDER HEIDEN ET AL., SCIENCE, vol. 324, no. 5930, 2009, pages 1029 - 1033
WARBURG, SCIENCE, vol. 124, no. 3215, 1956, pages 269 - 270
WARBURG ET AL.: "Metabolism of leukocytes]. Zeitschriftfur Naturforschung. TeilB: Chemie", BIOCHEMIE, BIOPHYSIK, BIOLOGIE, vol. 13B, no. 8, 1958, pages 515 - 516
"Current Protocols in Molecular Biology", December 2008, JOHN WILEY & SONS, article "Current Protocols in Immunology, Current Protocols in Protein Science, and Current Protocols in Cell Biology"
ZGODKA ET AL., MICROBIOLOGY, vol. 147, 2001, pages 1955 - 1959
SULLIVAN ET AL., CANCER CHEMOMETABOLIC REPROGRAMMING THERAPY AND PHARMACOLOGY, vol. 21, no. 1, 1988, pages 78 - 84
CHAMGAJEWSKI, JOURNAL OF IMMUNOLOGY, vol. 174, no. 8, 2005, pages 4670 - 4677
POWELLZHENG, CURR OPIN INVESTIG DRUGS, vol. 7, no. 11, 2006, pages 1002 - 1007
DARMAUN ET AL.: "Phenylbutyrate-induce glutamine depletion in humans: effect on leucine metabolism", GLUTAMINE DEPLETION AND PROTEIN CATABOLISM, AM. PHYSIOL. SOC., 1998, pages E801 - E807
VAROQUI ET AL., J. BIOL. CHEM., vol. 275, no. 6, 2000, pages 4049 - 4054
PAWLIK ET AL., AM. J. PHYSIOL. GASTROINTEST. LIVER PHYSIOL.,, vol. 278, 2000, pages G532 - G541
ACEVEDO ET AL., TETRAHEDRON, vol. 57, 2001, pages 6353 - 6359
SRIKANTH ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY, 2002
CHEN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 3054 - 3057
KARLIN AND ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 22264 - 2268
KARLIN AND ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5877
ALTSCHUL ET AL., J. MOT BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
SHAH ET AL., CURR. OPIN. DRUG DISCOV. DEVEL., vol. 13, 2010, pages 466 - 80
HART ET AL., MOV. DISORD., vol. 24, 2009, pages 647 - 54
SIDMAN ET AL., BIOPOLYMERS, vol. 22, 1983, pages 547
LANGER ET AL., J. BIOMED. MATER. RES., vol. 15, 1981, pages 167
LANGER, CHEM. TECH., vol. 12, 1982, pages 98
EPSTEIN ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 82, 1985, pages 3688
HWANG ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 77, 1980, pages 4030
CHAPMAN, ADV. DRUG DELIV. REV., vol. 54, 2002, pages 531 - 545
CUNNINGHAM-RUNDLES, J. IMMUNOL. METH., vol. 152, 1992, pages 177 - 190
F. C. KULL ET AL., APPLIED MICROBIOLOGY, vol. 9, 1961, pages 538
EL-MIR ET AL., THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 275, no. 1, 2000, pages 223 - 228
THOMAS ET AL., BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 438, no. 2, 2013, pages 243 - 248
CORRY ET AL., TRANSPLANTATION, vol. 16, no. 4, 1973, pages 343 - 350
OBERHUBE ET AL., J VIS EXP, vol. 92, 2014, pages e50753
SAYEGHCARPENTER, THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 351, no. 26, 2004, pages 2761 - 2766
CRUTCHLOWBLOOM, CLINICAL JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY: CJASN, vol. 2, no. 2, 2007, pages 343 - 355
HOORN ET AL., JOURNAL OF NEPHROLOGY, vol. 25, no. 3, 2012, pages 269 - 275
ARNOLD ET AL., AM J TRANSPLANT, vol. 13, no. 9, 2013, pages 2426 - 2432
GUBA ET AL., TRANSPLANTATION, vol. 77, no. 12, 2004, pages 1777 - 1782
ROODNAT ET AL., TRANSPLANTATION, vol. 98, no. 1, 2014, pages 47 - 53
WU ET AL., TRANSPL IMMUNOL, vol. 27, no. 1, 2012, pages 30 - 38
YANGCHI, SEMINARS IN IMMUNOLOGY, vol. 24, no. 6, 2012, pages 421 - 428
WAICKMANPOWELL, IMMUNOLOGICAL REVIEWS, vol. 249, no. 1, 2012, pages 43 - 58
POLLIZZIPOWELL, NATURE REVIEWS. IMMUNOLOGY, vol. 14, no. 7, 2014, pages 435 - 446
POWELL ET AL., COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY, vol. 78, 2013, pages 125 - 130
PEARCE ET AL., SCIENCE, vol. 342, no. 6155, 2013, pages 1242454
JONESTHOMPSON, IMMUNITY, vol. 27, no. 2, 2007, pages 173 - 178
WARBURG ET AL.: "Metabolism of leukocytes]. Zeitschrift fur Naturforschung", TEILB: CHEMIE, BIOCHEMIE, BIOPHYSIK, BIOLOGIE, vol. 13B, no. 8, 1958, pages 515 - 516
CHAM ET AL., EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 38, no. 9, 2008, pages 2438 - 2450
JACOBS ET AL., JOURNAL OF IMMUNOLOGY, vol. 180, no. 7, 2008, pages 4476 - 4486
CARR ET AL., JOURNAL OF IMMUNOLOGY, vol. 185, no. 2, 2010, pages 1037 - 1044
MICHALEK ET AL., JOURNAL OF IMMUNOLOGY, vol. 186, no. 6, 2011, pages 3299 - 330322
SHI ET AL., THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 208, no. 7, 2011, pages 1367 - 1376
MACINTYRE ET AL., CELL METAB, vol. 20, no. 1, 2014, pages 61 - 72
NAKAYA ET AL., IMMUNITY, vol. 40, no. 5, 2014, pages 692 - 705
ROWE ET AL., NATURE MEDICINE, vol. 19, no. 4, 2013, pages 488 - 493
CHENG ET AL., CANCER RESEARCH, vol. 72, no. 10, 2012, pages 2634 - 2644
YAMASAKI ET AL., NATURE REVIEWS. UROLOGY, vol. 8, no. 3, 2011, pages 165 - 171
BUZZAI ET AL., CANCER RESEARCH, vol. 67, no. 14, 2007, pages 6745 - 6752
CHEONG ET AL., MOLECULAR CANCER THERAPEUTICS, vol. 10, no. 12, 2011, pages 2350 - 2362
CHENG ET AL., BRITISH JOURNAL OF CANCER, vol. 111, no. 1, 2014, pages 85 - 93
MACLVER ET AL., ANNUAL REVIEW OF IMMUNOLOGY, vol. 31, 2013, pages 259 - 283
THOMAS ET AL., BIOCHEMICAL AND BBIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 443, no. 1, 2014, pages 32 - 36
GORSKIWASIK, IMMUNOLOGY, vol. 71, no. 1, 1990, pages 148 - 150
FRANCIOSI ET AL., PLOS ONE, vol. 8, no. 8, 2013, pages e71583
RAEZ ET AL., CANCER CHEMOMETABOLIC REPROGRAMMING THERAPY AND PHARMACOLOGY, vol. 71, no. 2, 2013, pages 523 - 530
TSILIDIS ET AL., DIABETES CARE, vol. 37, no. 9, 2014, pages 2522 - 2532
CRUTCHLOWBLOOM, CLINICAL JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY:CJASN, vol. 2, no. 2, 2007, pages 343 - 355
CHAMBERS ET AL., J VIROL, vol. 84, no. 4, 2010, pages 1867 - 1873
WILLEMS ET AL., BLOOD, vol. 122, no. 20, 2013, pages 3521 - 3532
LIM ET AL., CANCER RESEARCH, vol. 74, no. 13, 2014, pages 3535 - 3545
ZHENG ET AL., JOURNAL OF IMMUNOLOGY, vol. 183, no. 10, 2009, pages 6095 - 6101
ODERUP ET AL., TRANSPLANTATION, vol. 82, no. 11, 2006, pages 1493 - 1500
PILON ET AL., AMERICAN JOURNAL OF TRANSPLANTATION: OFFICIAL JOURNAL OF THE AMERICAN SOCIETY OF TRANSPLANTATION AND THE AMERICAN SOCIETY OF TRANSPLANT SURGEONS, vol. 14, no. 12, 2014, pages 2874 - 2882
BESTARD ET AL., TRANSPLANT INTERNATIONAL: OFFICIAL JOURNAL OF THE EUROPEAN SOCIETY FOR ORGAN TRANSPLANTATION, vol. 24, no. 5, 2011, pages 451 - 460
DELGOFFE ET AL., NATURE IMMUNOLOGY, vol. 12, no. 4, 2011, pages 295 - 303
WANG ET AL., IMMUNITY, vol. 35, no. 6, 2011, pages 871 - 882
DELGOFFE ET AL., IMMUNITY, vol. 30, no. 6, pages 832 - 844
ABDELMALEK ET AL., AMERICAN JOURNAL OF TRANSPLANTATION: OFFICIAL JOURNAL OF THE AMERICAN SOCIETY OF TRANSPLANTATION AND THE AMERICAN SOCIETY OF TRANSPLANT, vol. 12, no. 3, 2012, pages 694 - 705
LO ET AL., CURRENT OPINION IN ORGAN TRANSPLANTATION, vol. 19, no. 4, 2014, pages 363 - 371
SATAKE ET AL., PLOS ONE, vol. 9, no. 3, 2014, pages e9288854
SENGUPTA ET AL., MOLECULAR CELL, vol. 40, no. 2, 2010, pages 310 - 322
CSIBI ET AL., CELL, vol. 153, no. 4, 2013, pages 840 - 854
See also references of EP 3328376A4
Attorney, Agent or Firm:
FAZZOLARE, David A. (US)
Download PDF:
Claims:
THAT WHICH IS CLAIMED:

1. A method for treating a subject having 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, and/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 method comprising administering to the subject at least one, at least two, or at least three metabolic reprogramming agent 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 in an amount effective to treat the condition, disease, or disorder.

2 The method of claim 1, wherein:

(i) at least one metabolic reprogramming agent decreases glutamine metabolism;

(ii) at least one metabolic reprogramming agent decreases glycolysis; and/or (ii) at least one metabolic reprogramming agent increases fatty acid oxidation.

3. The method of claim 2, wherein the at least one metabolic

reprogramming agent that decreases glutamine metabolism is:

(i) a glutamine antagonist;

(ii) a glutamine analog that interferes with a glutamine metabolic pathway;

(iii) 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, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4- oxo-L-norvaline (L-DONV);

(iv) a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway; or

(v) a prodrug of acivicin, azaserine, DON, and L-DONV; and combinations thereof.

4. The method of claim 2, wherein the at least one metabolic reprogramming agent that decreases glycolysis is:

(i) a glucose analog that inhibits hexokinase; or

(ii) 2-deoxy-D-glucose (2-DG); and combinations thereof

5. The method of claim 2, wherein the at least one metabolic reprogramming agent that increases fatty acid oxidation is:

(i) an activator of 5' AMP-activated protein kinase (AMPK) activity; or

(ii) metformin; and combinations thereof.

6. The method of claim 1, wherein the condition, disease, or disorder is selected from the group consisting of an immune disorder, transplant rejection, graft versus host disease, inflammation, CNS or brain inflammation, a pathology due to or associated with CNS inflammation due to an infection, a pathology due to or associated with CNS inflammation not involving an infection, and a

neurodegenerative disorder.

7. The method of claim 1, wherein the condition, disease, or disorder is selected from the group consisting of acute respiratory distress syndrome (ARDS), allograft rejection during cell, tissue, or organ transplantation, Alzheimer's Disease, amyotrophic lateral sclerotis (ALS), arthritis, asthma, cerebral malaria, lupus, neuromyelitis optica, Parkinson's Disease, pneumonitis, pulmonary fibrosis

8. The method of claim 1, wherein the metabolically reprogrammed cells are selected from the group consisting of immune cells and neuronal cells.

9. 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, at least two, or at least three metabolic pathways selected from the group consisting of glutamine metabolism, glycolysis, and fatty acid synthesis, and a pharmaceutically acceptable carrier, diluent, or excipient.

10. The composition of claim 9, wherein: (i) at least one metabolic reprogramming agent decreases glutamine metabolism;

(ii) at least one metabolic reprogramming agent decreases glycolysis; and/or (ii) at least one metabolic reprogramming agent increases fatty acid oxidation.

11. The method of claim 10, wherein the at least one metabolic reprogramming agent that decreases glutamine metabolism is:

(i) a glutamine antagonist;

(ii) a glutamine analog that interferes with a glutamine metabolic pathway;

(iii) 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, and 6-diazo-5-oxo-norleucine (DON), and 5-diazo-4- oxo-L-norvaline (L-DONV);

(iv) a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway; or

(v) a prodrug of acivicin, azaserine, DON, and L-DONV; and combinations thereof.

12. The method of claim 10, wherein the at least one metabolic reprogramming agent that decreases glycolysis is:

(i) a glucose analog that inhibits hexokinase; or

(ii) 2-deoxy-D-glucose (2-DG); and combinations thereof

13. The method of claim 10, wherein the at least one metabolic reprogramming agent that increases fatty acid oxidation is:

(i) an activator of 5' AMP-activated protein kinase (AMPK) activity; or

(ii) metformin; and combinations thereof.

14. The composition of claim 9 further comprising an additional therapeutic agent selected from the group consisting of an immunotherapeutic agent, an immunosuppressant agent, a radiotherapeutic agent, an anti-inflammatory agent, and a neuroprotective agent.

Description:
METHODS AND COMPOSITIONS FOR TREATING METABOLIC REPROGRAMMING DISORDERS

CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of U.S. Provisional Application Nos.

62/199,381 and 62/199,566, both filed July 31, 2015, the contents of each are incorporated herein by reference in their entirety.

BACKGROUND

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. For example, cells under certain conditions may undergo a switch toward increased glycolysis and away from oxidative phosphorylation (OXPHOS). While glycolysis provides less adenosine triphosphate (ATP) than oxidative phosphorylation, it has been proposed that 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). This use of glycolysis in the presence of oxygen was first described by Otto Warburg in cancer cells (Warburg (1956) Science 124 (3215):269- 270) and was subsequently found to be important in activated T cells (Warburg et al. (1958) [Metabolism of leukocytes]. Zeitschrift fur Naturforschung. Teil B: Chemie, Biochemie, Biophysik, Biologie 13B (8):515-516). These metabolically

reprogrammed cells depend on the increased activity of certain metabolic pathways, such as pathways involved in glutamine metabolism, glycolysis, and fatty acid synthesis. However, specific inhibitors of individual enzymes in these metabolic pathways alone have not proven effective because multiple points within each metabolic pathway are modulated as a cell's metabolism is reprogrammed to meet the extraordinarily large energy demands of the abnormal, harmful, or unhealthy state.

SUMMARY

The practice of the present invention will typically employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant nucleic acid (e.g., DNA) technology, immunology, and RNA interference (RNAi) which are within the skill of the art. Non- limiting descriptions of certain of these techniques are found in the following publications: Ausubel, F., et al, (eds.), Current Protocols in Molecular Biology, Current Protocols in Immunology, Current Protocols in Protein Science, and Current Protocols in Cell Biology, all John Wiley & Sons, N.Y., edition as of December 2008; Sambrook, Russell, and Sambrook, Molecular Cloning. A Laboratory Manual, 3 rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 2001 ; Harlow, E. and Lane, D., Antibodies— A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1988; Freshney, R. I., "Culture of Animal Cells, A Manual of Basic Technique", 5th ed., John Wiley & Sons, Hoboken, N.J., 2005. Non-limiting information regarding therapeutic agents and human diseases is found in Goodman and Gilman's The Pharmacological Basis of Therapeutics, 11th Ed., McGraw Hill, 2005, Katzung, B. (ed.) Basic and Clinical Pharmacology, McGraw-Hill/ Appleton & Lange 10 th ed. (2006) or 11th edition (July 2009). Non-limiting information regarding genes and genetic disorders is found in McKusick, V. A.: Mendelian Inheritance in Man. A Catalog of Human Genes and Genetic Disorders. Baltimore: Johns Hopkins University Press, 1998 (12th edition) or the more recent online database: Online Mendelian Inheritance in Man, OMIM™. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore, Md.) and National Center for Biotechnology Information, National Library of Medicine (Bethesda, Md.), as of May 1, 2010, World Wide Web URL: http://www.ncbi.nlm.nih.gov/omim/ and in Online Mendelian Inheritance in Animals (OMIA), a database of genes, inherited disorders and traits in animal species (other than human and mouse), at

http://omia. angis. org. au/ contact, shtml.

In an aspect, 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. In an aspect, 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.

In an aspect, 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.

In an aspect, the presently disclosed subject matter provides 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. In particular embodiments, the pharmaceutical composition further includes at least one additional therapeutic agent selected from the group consisting of cytotoxic agents, immunotherapeutic agents, immunosuppressant agents,

radiotherapeutic agents, anti-inflammatory agents, and neuroprotective agents.

In particular embodiments, the method or use 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.

In particular embodiments, the method or use comprises administering to the subject at least three metabolic reprogramming agents that each decrease the activity of a different 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.

In particular embodiments, at least one metabolic reprogramming agent decreases glutamine metabolism. In particular embodiments, at least one metabolic reprogramming agent is a glutamine antagonist. In particular embodiments, at least one metabolic reprogramming agent is a glutamine analog that interferes with a glutamine metabolic pathway. In particular embodiments, 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, and 6- diazo-5-oxo-norleucine (DON), and 5-diazo-4-oxo-L-norvaline (L-DONV). In particular embodiments, at least one metabolic reprogramming agent is a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway. In particular embodiments, at least one metabolic reprogramming agent is a prodrug of acivicin, azaserine, DON, and L-DONV.

In particular embodiments, at least one metabolic reprogramming agent decreases glycolysis. In particular embodiments, at least one metabolic

reprogramming agent is a glucose analog that inhibits hexokinase. In particular embodiments, at least one metabolic reprogramming agent is 2-deoxy-D-glucose (2- DG).

In particular embodiments, at least one metabolic reprogramming agent increases fatty acid oxidation. In particular embodiments, at least one metabolic reprogramming agent is an activator of 5' AMP-activated protein kinase (AMPK) activity. In particular embodiments, at least one metabolic reprogramming agent is metformin.

In particular embodiments, the condition, disease, or disorder is an immune disorder. In particular embodiments the metabolically reprogrammed cells comprise immune cells. In particular embodiments, the condition, disease, or disorder is a neurodegenerative disorder. In particular embodiments, the metabolically reprogrammed cells comprise neuronal cells. In particular embodiments, the condition, disease, or disorder is a pathology due to or associated with CNS inflammation due to an infection. In particular embodiments, the condition, disease, or disorder is cereberal malaria. In particular embodiments, the condition, disease, or disorder is a pathology due to or associated with CNS inflammation not involving an infection. In particular embodiments, the condition, disease, or disorder is amyotrophic lateral sclerosis (ALS). In particular embodiments, the condition, disease, or disorder is Alzheimer's Disease). In particular embodiments, the condition, disease, or disorder is Parkinson's Disease). In particular embodiments, the condition, disease, or disorder is neuromyelitis optica). In particular embodiments, the condition, disease, or disorder is ARDS. In particular embodiments, the condition, disease, or disorder is arthritis. In particular embodiments, the condition, disease, or disorder is asthma. In particular embodiments, the condition, disease, or disorder is allograft rejection during cell, tissue, or organ transplantation. In particular embodiments, the method for treating, preventing, or delaying a transplant rejection (e.g., allograft rejection, e.g., heart allograft rejection) further includes transplanting a cell, tissue, or organ into the subject. In particular embodiments, the cell, tissue, or organ allograft comprises a MHC mismatched allograft (e.g., complete MHC mismatch). In particular embodiments, the method further includes administering an effective amount of an immunosuppressant agent to the subject. In particular embodiments, the condition, disease, or disorder is cerebral malaria. In particular embodiments, the condition, disease, or disorder is lupus. In particular embodiments, the condition, disease, or disorder is pneumonitis. In particular embodiments, the condition, disease, or disorder is pulmonary fibrosis.

In particular embodiments, the condition, disease, or disorder is not an immune disorder comprising multiple sclerosis. In particular embodiments, the condition, disease, or disorder is not a neurodegenerative disorder selected from the group consisting of Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Huntington's disease, stroke, and transient ischemic brain injury.

In an aspect, the presently disclosed subject matter provides a method for the treatment of ARDS 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 ARDS in the subject. In an aspect, 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 ARDS in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of arthritis 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 arthritis in the subject. In an aspect, 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 arthritis in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of asthma 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 asthma in the subject. In an aspect, 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 asthma in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of amyotrophic lateral sclerosis (ALS) 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 ALS in the subject. In an aspect, 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 ALS in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of Alzheimer's Disease 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 Alzheimer's Disease in the subject. In an aspect, 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 Alzheimer's Disease in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of cerebral malaria 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 cerebral malaria in the subject. In an aspect, 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 cerebral malaria in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for preventing or delaying allograft rejection during cell, tissue, or organ transplantation in a subject in need thereof, the method comprising administering to a subject about to undergo, undergoing, or having undergone a cell, tissue, or organ transplantation at least one metabolic reprogramming agent that decreases glutamine metabolism in an amount effective to prevent or delay allograft rejection of the cell, tissue, or organ transplanted in the subject. In particular embodiments, the method includes comprising transplanting a cell, tissue, or organ into the subject. In particular embodiments, the method further includes administering an immunosuppressant agent to the subject. In an aspect, 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, prevent, or delay an allograft rejection in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides method for the treatment of lupus 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 lupus in the subject. In an aspect, 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 lupus in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of neuromyeletis optica 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 neuromyeletis optica in the subject. In an aspect, 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 neuromyeletis optica in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of Parkinson's Disease 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 Parkinson's Disease in the subject. In an aspect, 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 Parkinson's Disease in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of pneumonitis 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 pneumonitis in the subject. In an aspect, 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 pneumonitis in a subject in need thereof.

In an aspect, the presently disclosed subject matter provides a method for the treatment of pulmonary fibrosis 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 pulmonary fibrosis in the subject. In an aspect, 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 pulmonary fibrosis in a subject in need thereof.

In particular embodiments, at least one metabolic reprogramming agent that decreases glutamine metabolism comprises a glutamine antagonist. In particular embodiments, at least one metabolic reprogramming agent that decreases glutamine metabolism comprises a glutamine analog that interferes with a glutamine metabolic pathway. In particular embodiments, at least one metabolic reprogramming agent that decreases glutamine metabolism is selected from the group consisting of acivicin, azaserine, and DON, and L-DONV. In particular embodiments, at least one metabolic reprogramming agent that decreases glutamine metabolism is a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway. In particular embodiments, at least one metabolic reprogramming agent that decreases glutamine metabolism is a prodrug of acivicin (L-(alpha S, 5S)-alpha-amino-3-chloro-4,5- dihydro-5-isoxazoleacetic acid), azaserine, and 6-diazo-5-oxo-norleucine (DON).

In particular embodiments, the methods of treatment (e.g., ARDS, arthritis, ALS, Alzheimer's Disease, cerebral malaria, allograft rejection, lupus, neuromyeletis optica, Parkinson's Disease, pneumonitis, pulmonary fibrosis, etc.) or uses further include or involve administering to the subject an effective amount of at least one metabolic reprogramming agent that decreases glycolysis. In particular embodiments, at least one metabolic reprogramming agent that decreases glycolysis is a glucose analog that inhibits hexokinase. In particular embodiments, at least one metabolic reprogramming agent that decreases glycolysis is 2-deoxy-D-glucose (2-DG).

In particular embodiments, the methods of treatment (e.g., ARDS, arthritis, ALS, Alzheimer's Disease, cerebral malaria, allograft rejection, lupus, neuromyeletis optica, Parkinson's Disease, pneumonitis, pulmonary fibrosis, etc.) or uses further include or involve administering to the subject an effective amount of at least one metabolic reprogramming agent that increases fatty acid oxidation. In particular embodiments, at least one metabolic reprogramming agent that increases fatty acid oxidation is an activator of 5' AMP-activated protein kinase (AMPK) activity. In particular embodiments, at least one metabolic reprogramming agent that increases fatty acid oxidation is metformin.

Certain aspects of the presently disclosed subject matter having been stated hereinabove, which are addressed in whole or in part by the presently disclosed subject matter, other aspects will become evident as the description proceeds when taken in connection with the accompanying Examples and Drawings as best described herein below.

BRIEF DESCRIPTION OF THE DRAWINGS

Having thus described the presently disclosed subject matter in general terms, reference will now be made to the accompanying Figures, which are not necessarily drawn to scale, and wherein:

FIG. 1A shows that metabolic reprogramming is a critical component of immune cell differentiation and function;

FIG. IB shows the targeting of glutamine metabolism to regulate immune effector function;

FIG. 1C, FIG. ID, and FIG. IE show that the presently disclosed metabolic reprogramming agents suppress antigen-specific effector CD4 + T cells proliferation and function. Thyl .2 + WT B6 mice were treated with vehicle, metabolic

reprogramming therapy with at least two metabolic reprogramming agents (e.g., 2DG + Metformin), or metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON). DON was found to be a very potent inhibitor of CD4 + T cell effector function.

FIG. 2A, FIG. 2B, and FIG. 2C show that the presently disclosed metabolic reprogramming agents suppress antigen-specific CD8 + T cells responses in vivo. Thy 1.2 + WT B6 mice were treated with vehicle, metabolic reprogramming therapy with at least two metabolic reprogramming agents (e.g., 2DG + Metformin), or metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON). DON was found to be a very potent inhibitor of CD8 + T cell effector function;

FIG. 3 shows that DON prevents transplant rejection and prolongs skin graft survival. Mouse full thickness skin from Balb/c mice were transplanted onto B6 mice and the mice were not treated with metabolic reprogramming therapy (No TX), or treated with metabolic reprogramming therapy with at least two metabolic reprogramming agents (e.g., 2DG + Metformin), metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON), or metabolic reprogramming with at least one metabolic reprogramming agent (e.g., DON);

FIG. 4 shows that DON prevents heart transplant rejection. Hearts from Balb/c mice were transplanted into B6 mice and the mice were not treated with metabolic reprogramming therapy (vehicle) or treated with metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON) for 100 days;

FIG. 5 shows that the metabolic reprogramming agents increase the relative frequency of regulatory T cells (Tregs) in vivo. Mice were not treated with metabolic reprogramming therapy (vehicle), or treated with metabolic reprogramming therapy with at least two metabolic reprogramming agents (e.g., 2DG + Metformin), or metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON);

FIG. 6 shows reprogramming metabolism in asthma with mock, no treatment, or metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON);

FIG. 7 shows that metabolic reprogramming therapy with at least one metabolic reprogramming agent (e.g., DON) treats arthritis in a mouse model;

FIG. 8 shows that metabolic reprogramming therapy with at least one metabolic reprogramming agent (e.g., DON) treats pneumonitis in a mouse model;

FIG .9A, FIG. 9B, FIG. 9C and FIG. 9D show that metabolic reprogramming therapy with metabolic reprogramming agent 2-DG combined with metabolic reprogramming agent metformin inhibits T cell responses through suppression of glycolysis. FIG. 9A and FIG. 9B show ECAR and OCR of resting CD4 + cells measured in real time under basal conditions and in response to anti-CD3/CD28 (anti- CD3, 2 μg/ml; anti-CD28, 2 μg/ml) with or without the presence of individual or combination of metabolic reprogramming agents (2-DG, 10 mM; Metformin, 50 mM). Bar graphs display data of ECAR and OCR measured at the endpoint of the experiment (205 min). Data are shown as mean ± SEM of 5 measurements. FIG. 9C and FIG. 9D show that naive splenocytes labeled with cell proliferation dye eFluor 670 were stimulated with anti-CD3 in in the presence of media control, 2-DG alone, metformin alone or 2-DG + metformin (2-DG, 0.6mM; Metformin, ImM). FIG. 9C shows that 24-hour IFN-γ secretion to supernatants was interrogated by enzyme- linked immunosorbent assay (ELISA). Data are shown as mean ± SEM of three independent samples. FIG. 9D shows 72-hour eFluor dilution of CD4 + and CD8 + T cells, n.s., not significant, *p<0.05, **p < 0.01, ****p < 0.0001 (Student's i test). Data are representative of at least two independent experiments;

FIG. 10A, FIG. 10B, and FIG. IOC show that the combined inhibition of at least two metabolic pathways (e.g., glycolysis and glutaminolysis) profoundly suppresses T cell responses. FIG. 10A shows the glutaminase activity of CD4 + T cells cultured for 24 hours in different conditions (anti-CD3, 2μg/ml; anti-CD28,

2μg/ml; DON, 5 μΜ; 2-DG, 0.6 mM; Metformin, 1 mM). Data are shown as mean ± SEM of three independent experiments. FIG. 10B and FIG. IOC show naive WT C57BL/6 splenocytes labeled with eFluor 670 and stimulated with anti-CD3 in medium containing indicated metabolic reprogramming agents (DON, 5μΜ; 2-DG, 0.6mM; Metformin, ImM). FIG. 10B shows 24-hour IFN-γ secretion to supernatants as interrogated by ELISA. Data are shown as mean ± SEM of three independent samples. FIG. IOC shows proliferation of CD4 + and CD8 + T cells at 72h measured by dilution of eFluor 670. n.s., not significant, **p < 0.01, ***p < 0.001 ,****p < 0.0001 (Student's t test). Data are representative of at least two independent experiments;

FIG. 11 shows the cell viability of T cells in different culture conditions.

Splenocytes from C57BL/6 mice were stimulated with anti-CD3 (1 μg/ml) in medium containing indicated metabolic reprogramming agents (DON, 5 μΜ; 2DG, 0.6 mM; Metformin, 1 mM) or media control. Cell viability was determined by 7- aminoactinomycin D (7-AAD) exclusion at 24 h by flow cytometry. Data are merged results of CD4 + and CD8 + cells and are shown as mean ± SEM. n.s., not significant (ANOVA). Data are representative of three independent experiments;

FIG. 12A and FIG. 12B show that DON does not affect ECAR and OCR of activated T cells. ECAR (FIG. 12A) and OCR (FIG. 12B) of resting CD4 + cells measured in real time under basal conditions and in response to anti-CD3/CD28 (anti- CD3, 2 μg/ml; anti-CD28, 2 μg/ml) with or without the presence of individual or combined metabolic reprogramming agents (2-DG, 10 mM; Metformin, 50 mM; DON, 30 μΜ). Bar graphs display ECAR and OCR data measured at the endpoint of the experiment (205 min). All data are shown as mean ± SEM of five measurements. n.s., not significant (Student's t test). Data are representative of two independent experiments;

FIG. 13 A, FIG. 13B, FIG. 13C, and FIG. 13D show that metabolic reprogramming agents suppress the proliferation and function of antigen-specific CD4 T cells while increasing the relative frequency of Tregs in vivo. OT-II

(Thyl . r ) CD4 + T cells were adoptively transferred into WT (Thy 1.2 ) recipient mice. The recipients were infected with OVA-expressing vaccinia virus and treated with vehicle, 2-DG+Metformin or 2-DG+metformin+DON (2-DG, 500 mg/kg once daily; metformin, 150 mg/kg once daily; DON, 1.6 mg/kg once every other day) for 3 days. Splenocytes from the recipients were harvested at day 4 to interrogate the frequency of antigen-specific T cells and regulatory T cells. The percentage of Thy 1.1 + cells relative to CD4 + cells were analyzed by flow cytometry {left) and plotted as cumulative data {right) (FIG. 13 A). OT-II cells were re-challenged with OVA peptide class II (10 μg/ml) ex vivo for 24 hours (FIG. 13B). The supernatants were interrogated for IFN-γ production by ELISA. Data are mean ± SEM (n=6).

Percentage of Foxp3+ cells relative to CD4+ cells {left) and plotted as cumulative data (right) (FIG. 13C). The ratio of OVA-specific Foxp3+ T cells: Effector cells (FIG. 13D). Each symbol represents an individual mouse. Horizontal lines indicate mean± SEM. as., not significant, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (Student's t test). Data are representative of more than three independent

experiments;

FIG. 14A, FIG. 14B, FIG. 14C, and FIG. 14D show that metabolic reprogramming agents suppress antigen-specific CD8 + T cells responses in vivo. For FIG. 14A, FIG. 14B, and FIG. 14C, OT-I (Thyl . l + ) CD8 + T cells were adoptively transferred into WT (Thyl .2 + ) recipient mice. The hosts were infected with vaccinia- OVA and treated with vehicle, metabolic reprogramming therapy with at least two metabolic reprogramming agents (e.g., 2-DG+metformin) or metabolic

reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2- DG + metformin + DON) for 5 days. Host splenocytes were harvested at day 6 to interrogate the proliferation and function of antigen-specific CD8 + T cells. Percent Thy 1.1 cells relative to CD8 cells were analyzed by flow cytometry {left) and plotted as cumulative data {right) (FIG. 14A). Percent IFN-y-producing cells relative to CD8 + cells {left) and plotted as cumulative data {right) (FIG. 14B). OT-I cells were re-challenged with OVA peptide class I (ΙΟμ/ml) ex vivo for 24 hours (FIG. 14C). The supernatants were interrogated for IFN-γ production by ELISA. Data are mean ± SEM (n=5 ~6). The ability of metabolic reprogramming agents to suppress endogenous effector CD8 + T cells development was assessed with an in vivo CTL assay (FIG. 14D). Percent of specific killing was determined at 10 h after transferring target cells. Data are mean ± SEM (n=3mice/group). Each symbol represents an individual mouse. Horizontal lines indicate mean± SEM. n.s., not significant, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (Student's t test). All data are representative of more than three independent experiments; and

FIG. 15 A, FIG. 15B, FIG. 15C, FIG. 15D, FIG. 15E, FIG. 15F, FIG. 15G, FIG. 15H, FIG. 151, and FIG. 15J show that metabolic reprogramming agents promote allograft survival. FIG. 15A shows Balb/c to C57BL/6 skin graft survival, as monitored daily by assessment of macroscopic signs of rejection. FIG. 15B, FIG. 15C, and FIG. 15D show representative photomicrographs of skin graft histology (hematoxylin and eosin (H&E) staining) on post-transplant day 7 under an optical microscope (outlet, X100; inlet: X200). FIG. 15E shows a representative photograph of Balb/c skin graft at day 40. FIG. 15F shows Balb/c to C57BL/6 heart graft survival, as monitored daily by palpation of heart beating. FIG. 15G, FIG. 15H, FIG. 151, and FIG. 15J show representative photomicrographs of cardiac graft histology (H&E staining) on the indicated post-transplant day under an optical microscope (outlet, X200; inlet, X400). The metabolic reprogramming treatment was started from the day of graft (day 0) until rejection in both models. The dosages of all drugs were the same as described in the FIG. 13 brief description. **p < 0.01,****p < 0.001 (Log-rank analysis). Data are representative of two independent experiments.

FIG. 16 shows the efficacy of the targeting of glutamine metabolism in a mouse model of asthma. Mice were sensitized to House Dust Mite antigen (HDM) in the absence of drug. Upon intratracheal rechallenge the mice were treated with vehicle or 25. In this model during the acute lung rechallenge 25 inhibited pathology, the recruitment/generation of Th2 cells and reduced the levels of HDM specific IgE.

FIG. 17A, FIG. 17B, and FIG. 17C show the efficacy of the targeting glutamine metabolism in a mouse model of Acute Respiratory Distress Syndrome (ARDS). Mice were challenged with Ips (FIG. 17A) to induce (ARDS) and treated with DON on day 2 & 4. Treatment promoted a more rapid recovery (weight gain) and less lung damage.

FIG. 18 shows the efficacy of the targeting of glutamine metabolism in a mouse model of Pulmonary Fibrosis. Mice were treated with intratracheal bleomycin to induce fibrosis. DON treated mice demonstrated markedly decreased lung damage as well as a decrease in the recruitment/generation of lung fibrocytes.

FIG. 19A and FIG. 19B show the efficacy of the targeting of glutamine metabolism to cure cerebral malaria. Mice were treated on day 5 post infection and on day 6 (12 hours before subsequent death) with DON every other day. The striking ability of DON to prevent death at such a late stage of disease highlights the robust ability of our approach in CNS inflammation.

FIG. 20 shows the efficacy of the targeting of glutamine metabolism in Neuromyelitis Optica. Auto-reactive T cells were adoptively transferred into mice treated with PBS or 25 and mean paralysis scores were measured.

FIG. 21 A and FIG. 21 B show the efficacy of the targeting of glutamine metabolism in Multiple Sclerosis. Animals were immunized for EAE

(C57BL/6+MOG35-55). Mice were treated with Vehicle or 25 either q.d. from the day of immunization (Day 0, left), or b.d. from the time of disease onset (EAE Disease Score >1, right).

FIG. 22 shows the results of the screening of prodrugs in vivo: Thy 1.1+ OT-1 cells were adoptively transferred into mice infected with vaccinia ova. The mice were treated with prodrugs or vehicle (V) and the Ova-specific response was monitored. In this case drugs PI, PI -2 and P3 effectively inhibited the T cell response.

FIG. 23A, FIG. 23B, and FIG. 23C illustrate the in vivo pharmacokinetics of

DON following i.v. administration of DON (1) and 14 in monkey plasma and cerebrospinal fluid (CSF). 1 and 14 were dosed in two pigtail macaques at 1.6 mg/kg equivalent of 1 via i.v. administration and plasma (0.25-6h) and CSF (30 min) concentrations of DON were evaluated via LC/MS. Relative to 1, 14 delivered substantially lower DON plasma concentration. The reverse was observed in CSF, where 14 delivered significantly higher DON CSF concentrations, achieving a 10-fold enhanced CSF to plasma ratio at 30 minute post dose. Note the bold numbers following the terms "DON", "DON prodrugs", "DON-based prodrugs", and the like, refer to particular compounds disclosed in then international PCT application directed to "Prodrugs of Glutamine Analogs," filed on July 31, 2016 (Attorney Docket No. 111232-00500), the entirety of which is incorporated herein by reference).

Specifically, other than 1, which refers to DON, the bold numbers refer to prodrugs (e.g., DON-prodrugs);

FIG. 24 illustrates species specific plasma stability of (14); 14 is stable in plasma of human, pig, dog and monkeys, but rapidly metabolized in mice;

FIG. 25 illustrates exemplary structures of DON and DON-based prodrugs 25, 9, 38 and 60; different N-amino acid promoeities (e,g, leucine, tryptophan) provide differential plasmas and microsomal stability;

FIG. 26A, FIG. 26B, FIG. 26C, and FIG. 26D illustrate the in vitro plasma stability of DON prodrugs 9, 25, 38 and 60. Metabolism occurs via removal of N- protecting group; both ethyl and isopropyl esters are stable in plasma of pigs and humans;

FIG. 27A, FIG. 27B, FIG. 27C, and FIG. 27D illustrates the in vitro liver microsomal stability of DON prodrugs 9, 25, 38 and 60; all prodrugs showed moderate-high stability in human and pig microsomes;

FIG. 28A, FIG. 28B, FIG. 28C, FIG. 28D, FIG. 28E, FIG. 28F, FIG. 28G, FIG. 28H, FIG. 281, and FIG. 28J illustrate the results of ex- vivo studies in whole human and pig blood of 9, 25, 38 and 60; DON prodrugs selectively deliver DON to PBMCs in both humans and pigs vs plasma; compared to DON, the PBMC/plasma ratio was enhanced 10-100+ fold;

FIG. 29A FIG. 29B, FIG. 29C, FIG. 29D and FIG. 29E illustrate the results of pig in vivo studies with DON prodrugs of 9, 25, 38 and 60; DON prodrugs selectively deliver DON to PBMCs vs plasma; compared to DON, the PBMC/plasma ratio was enhanced 6- to 10-fold;

FIG. 30A, FIG. 30B, and FIG. 30C illustrate the plasma stability of compound Methyl-POM 14 and its derivatives; and

FIG. 31 illustrate 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. The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee. DETAILED DESCRIPTION

The presently disclosed subject matter now will be described more fully hereinafter with reference to the accompanying Figures, in which some, but not all embodiments of the presently disclosed subj ect matter are shown. Like numbers refer to like elements throughout. The presently disclosed subject matter may be embodied in many different forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will satisfy applicable legal requirements. Indeed, many modifications and other embodiments of the presently disclosed subj ect matter set forth herein will come to mind to one skilled in the art to which the presently disclosed subject matter pertains having the benefit of the teachings presented in the foregoing descriptions and the associated Figures. Therefore, it is to be understood that the presently disclosed subject matter is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims.

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. It should be appreciated that 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).

The presently disclosed subject matter have demonstrated that certain of such conditions, diseases, and/or disorders, referred to herein as "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. In some instances, 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.

As used herein, 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. As used herein, 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.).

As used herein, 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. As used herein, 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.).

As used herein, 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. As used herein, 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.).

As used herein, 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. As used herein, 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. In some contexts, 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 compositions, methods, and agents for metabolic

reprogramming may provide a number of advantages over existing therapies, including without limitation, inhibition of effector T cells while enhancing regulatory T cells, promoting immune tolerance as opposed to inhibiting it, providing a positive effect on systemic metabolism (e.g., avoids conditions such as steroid-induced metabolic syndrome/diabetes), inhibiting the replication of CMV and other viruses and other complications of immunosuppression, and providing a robust therapeutic index as demonstrated by in vivo studies (e.g., due to cellular selectivity based upon metabolic demand). The presently disclosed subject matter now will be described more fully hereinafter with reference to the accompanying Figures, in which some, but not all embodiments of the inventions are shown. Like numbers refer to like elements throughout. The presently disclosed subject matter may be embodied in many different forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will satisfy applicable legal requirements. Indeed, many modifications and other embodiments of the presently disclosed subject matter set forth herein will come to mind to one skilled in the art to which the presently disclosed subject matter pertains having the benefit of the teachings presented in the foregoing descriptions and the associated Figures.

Therefore, it is to be understood that the presently disclosed subject matter is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. I. METHODS OF TREATMENT USING METABOLIC REPROGRAMMING AGENTS

In an aspect, 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.

In particular embodiments, the presently disclosed subject matter provides a method for treating a condition, disease, or disorder that involves metabolically reprogrammed cells whose activation, function, growth, proliferation, and/or survival depends on increased glutamine metabolic activity, the method comprising administering to the subject an effective amount of an agent that decrases glutamine metabolic activity.

In some aspects, 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.

In particular embodiments, the presently disclosed subject matter provides a method for treating a subject having a condition, disease, or disorder that involves aberrant and/or excessive glutamine metabolism, the method comprising

administering to the subject an effective amount of an agent that decreases glutamine metabolic activity.

In general, 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. The term "decrease" is meant to inhibit, suppress, attenuate, diminish, arrest, or stabilize a symptom of the condition, disease, or disorder. As used herein, 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.

In some embodiments, 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. In other embodiments, 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. In still other embodiments, the method comprises administering at least four, five, six, or more metabolic reprogramming agents to the subject.

The terms "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." Accordingly, 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. An animal may be a transgenic animal. In some embodiments, the subject is a human including, but not limited to, fetal, neonatal, infant, juvenile, and adult subjects. Further, a "subj ect" can include a patient afflicted with or suspected of being afflicted with a condition or disease.

A. IMMUNE DISORDERS

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 an immunosuppressant agent and/or an anti-inflammatory agent, for the treatment of an immune disorder (e.g., an autoimmune disorder). Accordingly, in some embodiments, the condition, disease, or disorder is an immune disorder. In such embodiments, the metabolically reprogrammed cells comprise immune cells.

Examples of immune 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, antigen-specific effector CD4+ T cells, antigen-specific effector CD8+ t cells, and regulatory T cells.

As used herein, the term "immune disorders" includes diseases involving the immune system that can include but not be limited to allergies, autoimmune diseases, immune complex diseases, immunodeficiency diseases and cancers of the immune system. Autoimmunity is the failure of an organism to recognize its own constituent parts (down to the sub-molecular levels) as "self, which results in an immune response against its own cells and tissues. Any disease that results from such an aberrant immune response is termed an autoimmune disease. An unwanted immune response may be, for example, immune responses associated with an autoimmune disorder, transplants, allergies, or inflammatory disorders.

Exemplary autoimmune diseases and disorders that may be treated with the presently disclosed methods, uses, and compositions comprising one or more metabolic reprogramming agents include, for example, inflammatory responses, such as inflammatory skin diseases, including psoriasis and dermatitis (e.g. atopic dermatitis); dermatomyositis; systemic scleroderma and sclerosis; responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis); respiratory distress syndrome (including adult respiratory distress syndrome; ARDS); dermatitis; meningitis; encephalitis; uveitis; colitis; glomerulonephritis; allergic conditions, such as eczema and asthma, and other conditions involving infiltration of T cells and chronic inflammatory responses; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE);

diabetes mellitus (e.g. Type I diabetes mellitus or insulin dependent diabetes mellitus); multiple sclerosis; Reynaud's syndrome; autoimmune thyroiditis; allergic encephalomyelitis; Sjogren's syndrome; juvenile onset diabetes; and immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison's disease); diseases involving leukocyte diapedesis; central nervous system (CNS) inflammatory disorder; multiple organ injury syndrome; hemolytic anemia (including, but not limited to cryoglobinemia or Coombs positive anemia); myasthenia gravis; antigen-antibody complex mediated diseases; anti-glomerular basement membrane disease;

antiphospholipid syndrome; allergic neuritis; Graves' disease; Lambert-Eaton myasthenic syndrome; pemphigoid bullous; pemphigus; autoimmune

polyendocrinopathies; Reiter's disease; stiff-man syndrome; Bechet disease; giant cell arteritis; immune complex nephritis; IgA nephropathy; IgM polyneuropathies;

immune thrombocytopenic purpura (ITP) or autoimmune thrombocytopenia and autoimmune hemolytic diseases, Hashimoto's thyroiditis, Wegener's granulomatosis, cold agglutinin disease associated with indolent lymphoma, acquired factor VIII inhibitors disease, etc.

The term "immune disorders" are diseases involving the immune system that can include but not be limited to allergies, autoimmune diseases, immune complex diseases, immunodeficiency diseases and cancers of the immune system. The term "autoimmune diseases" may include but not be limited to Acute disseminated encephalomyelitis, Addison's disease, Ankylosing spondylitisis, Antiphospholipid antibody syndrome, Aplastic anemia, Autoimmune hepatitis, Autoimmune

Oophoritis, Coeliac disease, Crohn's disease, Diabetes mellitus, Gestational pemphigoid, Goodpasture's syndrome, Grave's disease, Guillan-Barre syndrome, Hashimoto's disease, Idiopathic thrombocytopenic purpura, Lupus erythematosus, Multiple sclerosis, Myasthenia gravis, Opsoclonus myoclonus syndrome, Optic neuritis, Ord's thyroiditis, Pemphigus, Pernicious anemia, Polyarthritis, Primary biliary cirrhosis, Rheumatoid arthritis, Reiter's syndrome, Sjogren's syndrome, Takayasu's arteritis, Warm autoimmune hemolytic anemia, and Wegener's granulomatosis. The term "chronic inflammatory diseases" may include but not be limited to Tuberculosis, Chronic cholecystitis, Bronchiectasis, ulcerative colitis, silicosis and other pneumoconiosis as well as the above listed autoimmune diseases.

In some embodiments, the condition, disease, or disorder is ARDS.

Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of ARDS 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 ARDS in the subject.

In some embodiments, the condition, disease, or disorder is arthritis.

Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of arthritis 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 arthritis in the subject.

In some embodiments, the condition, disease, or disorder is asthma.

Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of asthma 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 asthma in the subject.

The presently disclosed methods, compositions, and agents can be used to treat or prevent transplant rejections.

In some embodiments, the condition, disease, or disorder is allograft rejection during cell, tissue, or organ transplantation. Accordingly, in an aspect the presently disclosed subject matter provides a method for preventing or delaying allograft rejection during cell, tissue, or organ transplantation in a subject in need thereof, the method comprising administering to a subject about to undergo, undergoing, or having undergone a cell, tissue, or organ transplantation at least one metabolic reprogramming agent that decreases glutamine metabolism in an amount effective to prevent or delay allograft rejection of the cell, tissue, or organ transplanted in the subject.

In particular embodiments, a method for preventing or delaying a heart allograft rejection comprises administering to a subject about to undergo, undergoing, or having undergone a heart allograft transplation an effective amount of at least one metabolic reprogramming agent, at least two metabolic reprogramming agents, or at least three metabolic reprogramming agents selected from the group consisting of a metabolic reprogramming agent that decreases activity of glutamine metabolism, a metabolic reprogramming agent that decreases activity of glycolysis, and a metabolic reprogramming agent that decreases activity of fatty acid synthesis, thereby preventing or delaying rejection of the heart allograft in the subject. In some embodiments, the subject receiving the heart allograft transplantation is a full MHC mismatch relative to the donor of the heart allograft. In some embodiments, the subject is administered at least one metabolic reprogramming agent to prevent or delay the heart allograft rejection. In some embodiments, the subject is administered at least two metabolic reprogramming agents to prevent or delay the heart allograft rejection. In some embodiments, the subject is administered at least three metabolic reprogramming agent to prevent or delay the heart allograft rejection.

In some embodiments, the condition, disease, or disorder is graft versus host disease (GVHD). Accordingly, in an aspect the presently disclosed subject matter provides a method for treating or preventing GVHD in a subject in need thereof, the method comprising administering to a subject about to undergo, undergoing, or having undergone a cell, tissue, or organ transplantation at least one metabolic reprogramming agent that decreases glutamine metabolism in an amount effective to treat or prevent GVHD in the subject.

In some embodiments, the condition, disease, or disorder is lupus.

Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of lupus 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 lupus in the subject.

In some embodiments, the condition, disease, or disorder is multiple sclerosis. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of multiple sclerosis 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 multiple sclerosis in the subject. In another aspect the presently disclosed subject matter provides a method for the prevention of multiple sclerosis 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 prevent multiple sclerosis in the subject. As used herein "prevention" in the context of multiple sclerosis includes preventing the onset of the disease, as well as progression and/or severity of the progression of the disease. In some embodiments, the prevention method includes selecting a subject at risk of developing multiple sclerosis. For example, it is believed that having a first-degree relative such as a parent or sibling with multiple sclerosis increases an individual's risk of developing the disease. As such, the method contemplates selecting such subject and administering at least one metabolic reprogramming agent to the subject to prevent the onset of multiple sclerosis in the subject. In some embodiments, the prevention method includes preventing the progress and/or severity of progression of multiple sclerosis. In such embodiments, a subject, for example one who has recently been diagnosed as having multiple sclerosis, can be administered the at least one metabolic reprogramming agent to prevent the disease from progressing.

In some embodiments, the condition, disease, or disorder is neuromyelitis optica. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of neuromyelitis optica 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 neuromyelitis optica in the subject.

In some embodiments, the condition, disease, or disorder is pneumonitis. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of pneumonitis 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 pneumonitis in the subject.

In some embodiments, the condition, disease, or disorder is pulmonary fibrosis. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment of pulmonary fibrosis 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 pulmonary fibrosis in the subject.

In some embodiments, the condition, disease, or disorder is not an immune disorder comprising multiple sclerosis.

B. NEURODEGENERATIVE DISORDERS

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 neuroprotective agent, a neurotrophic factor, a neuroregenerative agent, an immunosuppressant agent, and/or an anti -inflammatory agent, for the treatment of a neurodegenerative disorder. Accordingly, in some embodiments, the condition, disease, or disorder is a neurodegenerative disorder. In such embodiments, the metabolically reprogrammed cells comprise neuronal cells. Examples of neuronal 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, glial cells (e.g., microglial cells), and astrocytes.

A "neurodegenerative disorder" is a disease, disorder, or condition that is characterized by the progressive loss of the structure or function of neurons (e.g., degeneration or dysfunction of neurons or other neural cells). Such diseases, disorders, or conditions include, but are not limited to, glaucoma, and

neurodegenerative diseases, disorders, or conditions of the nervous systems, such as or associated with amyotrophic lateral sclerosis (ALS), trigeminal neuralgia, glossopharyngeal neuralgia, Bell's Palsy, myasthenia gravis, muscular dystrophy, progressive muscular atrophy, primary lateral sclerosis (PLS), pseudobulbar palsy, progressive bulbar palsy, spinal muscular atrophy, inherited muscular atrophy, invertebrate disk syndromes, cervical spondylosis, plexus disorders, thoracic outlet destruction syndromes, peripheral neuropathies, prophyria, Alzheimer's disease, Huntington's disease, Parkinson's disease, Parkinson' s-plus diseases, multiple system atrophy, progressive supranuclear palsy, corticobasal degeneration, demyelinating diseases, Guillain-Barre syndrome, multiple sclerosis, Charcot-Marie-Tooth disease, prion diseases, Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker syndrome (GSS), fatal familial insomnia (FFI), bovine spongiform encephalopathy (BSE), Pick's disease, and epilepsy,.

Other neurodegenerative diseases, disorders, or conditions of the nervous systems, such as or associated with alcoholism, Alexander's disease, Alper's disease, ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Canavan disease, Cockayne syndrome, diabetic neuropathy, frontotemporal lobar degeneration, Kennedy's disease, Krabbe's disease, neuroborreliosis, Machado- Joseph disease (Spinocerebellar ataxia type 3), wet or dry macular degeneration, Niemann Pick disease, Pelizaeus-Merzbacher Disease, photoreceptor degenerative diseases, such as retinitis pigmentosa and associated diseases, Refsum's disease,

Sandhoff s disease, Schilder's disease, subacute combined degeneration of spinal cord secondary to pernicious anemia, Spielmeyer-Vogt-Sjogren-Batten disease (also known as Batten disease), spinocerebellar ataxia (multiple types with varying characteristics), Steele-Richardson-Olszewski disease, and tabes dorsalis.

Several further aspects of the presently disclosed subject matter relate to correcting defects associated with a wide range of genetic brain diseases. For example, genetic brain diseases may include but are not limited to

Adrenoleukodystrophy, Agenesis of the Corpus Callosum, Aicardi Syndrome, Alpers' Disease. Alzheimer's Disease, Barth Syndrome, Batten Disease, CADASIL,

Cerebellar Degeneration, Fabry's Disease, Gerstmann-Straussler-Scheinker Disease, Huntington's Disease and other Triplet Repeat Disorders, Leigh's Disease, Lesch- Nyhan Syndrome, Menkes Disease, Mitochondrial Myopathies and NINDS

Colpocephaly.

In other embodiments, the neurodegenerative disease, disorder, or condition is or is associated with a disease, disorder, or condition of the nervous system selected from the group consisting of amyotrophic lateral sclerosis (ALS), trigeminal neuralgia, glossopharyngeal neuralgia, Bell's Palsy, myasthenia gravis, muscular dystrophy, progressive muscular atrophy, primary lateral sclerosis (PLS),

pseudobulbar palsy, progressive bulbar palsy, spinal muscular atrophy, inherited muscular atrophy, invertebrate disk syndromes, cervical spondylosis, plexus disorders, thoracic outlet destruction syndromes, peripheral neuropathies, prophyria, Alzheimer's disease, Huntington's disease, Parkinson's disease, Parkinson's-plus diseases, multiple system atrophy, progressive supranuclear palsy, corticobasal degeneration, demyelinating diseases, Guillain-Barre syndrome, multiple sclerosis, Charcot-Marie-Tooth disease, prion diseases, Creutzfeldt- Jakob disease, Gerstmann- Straussler-Scheinker syndrome (GSS), fatal familial insomnia (FFI), bovine spongiform encephalopathy (BSE), Pick's disease, epilepsy, alcoholism, Alexander's disease, Alper's disease, ataxia telangiectasia, Batten disease, Canavan disease, Cockayne syndrome, diabetic neuropathy, frontotemporal lobar degeneration, Kennedy's disease, Krabbe's disease, neuroborreliosis, Machado- Joseph disease (Spinocerebellar ataxia type 3), wet or dry macular degeneration, Niemann Pick disease, Pelizaeus-Merzbacher Disease, photoreceptor degenerative diseases, Refsum's disease, Sandhof s disease, Schilder's disease, subacute combined degeneration of spinal cord secondary to pernicious anemia, Spielmeyer-Vogt- Sjogren-Batten disease (also known as Batten disease), spinocerebellar ataxia (multiple types with varying characteristics), Steele-Richardson-Olszewski disease, and tabes dorsalis.

In yet other embodiments, the neurodegenerative disease, disorder, or condition comprises one or more conditions that are secondary to a disease, disorder, condition, or therapy having a primary effect outside of the nervous system selected from the group consisting of: peripheral neuropathy or neuralgia caused by diabetes, cancer, hepatitis, kidney dysfunction, Colorado tick fever, diphtheria, leprosy, Lyme disease, polyarteritis nodosa, rheumatoid arthritis, sarcoidosis, Sjogren syndrome, syphilis, systemic lupus erythematosus, viral encephalitis, and amyloidosis. In some embodiments, the disease, condition, or disorder is viral encephalitis.

In other embodiments, the neurodegenerative disease, disorder, or condition is associated with pain selected from the group consisting of chronic pain, fibromyalgia, spinal pain, carpel tunnel syndrome, pain from cancer, arthritis, sciatica, headaches, pain from surgery, muscle spasms, back pain, visceral pain, pain from injury, dental pain, neuralgia, such as neurogenic or neuropathic pain, nerve inflammation or damage, shingles, herniated disc, a torn ligament, and diabetes.

In further embodiments, the neurodegenerative disease, disorder, or condition is associated with one or more injuries to the nervous system. In particular embodiments, the one or more injuries to the nervous system is related to nerve damage caused by exposure to one or more agents selected from the group consisting of toxic compounds, heavy metals, industrial solvents, drugs, chemotherapeutic agents, dapsone, cholesterol lowering drugs, heart or blood pressure medications, and metronidazole.

In more particular embodiments, the one or more injuries to the nervous system is related to nerve damage caused by one or more conditions selected from the group consisting of burn, wound, surgery, accidents, ischemia, prolonged exposure to cold temperature, stroke, intracranial hemorrhage, and cerebral hemorrhage.

In yet other embodiments, the neurodegenerative disease, disorder, or condition comprises a psychiatric disorder. In particular embodiments, the psychiatric disorder is selected from the group consisting of schizophrenia, delusional disorder, schizoaffective disorder, schizopheniform, shared psychotic disorder, psychosis, paranoid personality disorder, schizoid personality disorder, borderline personality disorder, anti-social personality disorder, narcissistic personality disorder, obsessive- compulsive disorder, delirium, mood disorders, bipolar disorder, depression, stress disorder, panic disorder, agoraphobia, social phobia, post-traumatic stress disorder, anxiety disorder, and impulse control disorders.

In some embodiments, the condition, disease, or disorder is Alzheimer's Disease. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment or prevention of Alzheimer's Disease 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 or prevent Alzheimer's Disease in the subject.

In some embodiments, the condition, disease, or disorder is amyotrophic lateral sclerosis (ALS). Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment or prevention of amyotrophic lateral sclerosis (ALS) 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 ALS in the subject.

In some embodiments, the condition, disease, or disorder is Parkinson's Disease. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment or prevention of Parkinson's Disease 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 or prevent Parkinson's Disease in the subject.

It is believed that certain of the presently disclosed metabolic reprogramming agents and compositions are particularly useful in the treatment of CNS inflammatory diseases. In particular, the data described in FIG. 23A, FIG. 23B, FIG. 23C, FIG. 30A, FIG. 30B, FIG. 30C and FIG. 31 demonstrate that certain metabolic reprogramming agents (e.g., prodrugs of glutamine analogs, e.g., DON prodrugs) effectively target and deliver DON to the brain, for example, achieving as much as a 10-fold enhanced CSF to plasma ratio at 30 minute post dosing. Accordingly, certain of the presently disclosed metabolic reprogramming agents are contemplated for use in the treatment of CNS inflammatory diseases, for example, due to their ability to selectively deliver metabolic reprogramming agents that decrease glutamine metabolism to the CNS.

In some embodiments, the condition, disease, or disorder is a pathology due to or associate with CNS inflammation due to an infection. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment or prevention of a pathology due to CNS inflammation due to an infection 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 or prevent the CNS inflammation due to the infection.

In some embodiments, the presently disclosed subject matter provides a method for the treatment or prevention of cerebral malaria, the method comprising administering to the subject an effective amount of at least one metabolic reprogramming agent that decreases glutamine metabolism in an amount effective to treat or prevent cerebral malaria. In some embodiments, at least one metabolic reprogramming agent treats or prevents the CNS inflammation and/or a symptom of the pathology. In some embodiments, the method includes selecting a subject with an infection (e.g., cerebral malaria, e.g., a subject who tests positive for a causative agent, e.g., infection with Plasmodium falciparum) for treatment with at least one metabolic repogramming agent.

In other embodiments, the condition, disease, or disorder is a pathology due to or associated with CNS inflammation not involving an infection. Accordingly, in an aspect the presently disclosed subject matter provides a method for the treatment or prevention of a pathology due to CNS inflammation non involving an infection 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 or prevent the CNS inflammation in the subject, thereby treating or preventing the pathology due to or associated with CNS inflammation in the subject. In some embodiments, the method includes selecting a subject having a pathology due to or associated with CNS inflammation not involving an infection. For example, a subject suffering from a pathology can be blood tested for one or more inflammatory markers (e.g., erythrocyte sedimention rate (ESR), C-reactive protein (CRP) and/or plasma viscosity (PV), and upon testing positive for one or more inflammatory markers, the subject can be tested for an infection. Subjects suffering from the pathology who test positive for inflammation and negative for an infection can be selected for treatment with at least one metabolic reprogramming agent to treat the pathology due to or associated with CNS inflammation not involving an infection. Examples of such pathologies include, but are not limited to amyotrophic lateral sclerosis, Alzheimer's Disease, and Parkinson's Disease.

In some embodiments, the condition, disease, or disorder is not a

neurodegenerative disorder selected from the group consisting of Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Huntington's disease, stroke, and transient ischemic brain injury.

II. METABOLIC REPROGRAMMING AGENTS

The presently disclosed subject matter contemplates the use of various agents in connection with the methods, uses, and compositions described herein. 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.

As used herein, "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. As used herein, "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. The term "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. As used herein, the term "modulator" comprises both inhibitors and activators of a metabolic pathway or target. For example,

"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).

As used herein, the phrase "modulation of a metabolic pathway" refers to modulation of activity of at least one component of the metabolic pathway. It is contemplated herein that 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.

The terms "decrease" , "reduced", "reduction", "decrease" or "inhibit" are all used herein generally to mean a decrease by a statistically significant amount.

However, for avoidance of doubt, ""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%. In one embodiment, 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.

Certain methods, compositions, and agents contemplated herein modulate an immune response and/or an inflammatory response. In the contexts of decreasing an immune response or inflammation, the methods, compositions, and agents contemplated herein can decrease the immune response or inflammation 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 or inflammation before employing the method, composition, and/or agent). In the contexts of increasing an immune response or inflammation, the methods, compositions, and agents contemplated herein can increase the immune response or inflammation 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 or inflammation before employing the method, composition, and/or agent).

Certain methods, compositions, and agents contemplated herein modulate neurodegeneration, dysfunction, and/or survival of neuronal cells. In the context of decreasing neurodegeneration and/or dysfunction of neuronal cells, the methods, compositions, and agents contemplated herein can decrease the neurodegeneration and/or dysfunction of neuronal 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 neurodegeneration and/or dysfunction of neuronal cells before employing the method, composition, and/or agent). In the context of increasing or promoting survival of neuronal cells, the methods, compositions, and agents contemplated herein can increase or promote survival of neuronal cells 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 obj ective measure of the survival of neuronal cells before employing the method, composition, and/or agent).

The term "statistically significant" or "significantly" refers to statistical significance and generally means a two standard deviation (2SD) below normal, or lower, concentration of the marker. The term 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.

As used more particularly herein in some contexts, "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. In other contexts, "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. In certain contexts, "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 a- ketoglutarate. As used herein, the term "glutamine metabolism" or "glutamine metabolic activity" refers to the chemical reactions, enzymes, and pathways involving glutamine. As used herein, the term "glutamine metabolic pathway" is a biochemical pathway that involves glutamine.

As can be envisioned by a person with skill in the art, a metabolic

reprogramming agent can modulate any of the chemical reactions, enzymes and/or pathways involving glutamine. In some embodiments, 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. In the context of decreasing 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).

In some embodiments, at least one metabolic reprogramming agent is a glutamine antagonist. As used herein, the term "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. When it is said that the 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. When it is said that 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.

In some embodiments, 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. It should be recognized that a compound that is a useful metabolic reprogramming agent may have two or more of these characteristics. For example, 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.

In some embodiments, at least one metabolic reprogramming agent can be an antagonist that inhibits the synthesis of glutamine. Examples of 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.

In some embodiments, at least one metabolic reprogramming agent can be a glutamine depleting enzyme. Examples of such 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.

In some embodiments, at least one metabolic reprogramming agent can be a compound that reacts with glutamine under intracellular conditions to form a non- glutamine product. An example of 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).

In some embodiments, at least one metabolic reprogramming agent can be an antagonist that inhibits glutamine uptake by cells. Examples of 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 Qt ?&., Am. J. Physiol. Gastrointest. Liver Physiol, 278:G532-G541 (2000)).

In some embodiments, at least one metabolic reprogramming agent can be a glutamine binding compound that reduces the biological availability of glutamine.

In some embodiments, at least one metabolic reprogramming agent can be a glutamine analog that interferes with a glutamine metabolic pathway. Examples of 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-d iazo-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, Zgodka et al, Microbiology, 147: 1955-1959 (2001)), (3S,4R)-3,4-dimethyl-L-glutamine, (3S,4R)-3,4-dimethyl-L- pyroglutamic acid (See, Acevedo et al, Tetrahedron., 57:6353-6359 (2001)), 1,5- N,N'-disubstituted-2-(substituted benzenesulphonyl) glutamamides (See, Srikanth et al., Bioorganic and Medicinal Chemistry, (2002)), or a mixture of any two or more of these. In some embodiments, 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).

In some embodiments, at least one metabolic reprogramming agent is a prodrug of a glutamine analog that interferes with a glutamine metabolic pathway. In some embodiments, 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" (Attorney Docket No. 111232-00403, filed concurrently herewith, and herein incorporated by reference in its entirety).

In some aspects, a prodrug of a glutamine antagonist, or a pharmaceutically acceptable salt or ester a (I):

(i);

wherein: X is selected from the group consisting of a bond, -0-, and -(CH 2 ) n - wherein n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8; Ri is selected from the group consisting of H and a first prodrug-forming moiety capable of forming a salt or an ester; and R2 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 R2; R 2 ' is selected from the group consisting of H, C1-C6 alkyl, substituted C1-C6 alkyl, or R 2 and R2' together form a ring structure comprising -C(=0)-G-C(=0)-, wherein G is selected from the group consisting of Ci-Cg alkylene, Ci-Cg heteroalkylene, Cs-Cg

cycloalkylene, C6-C12 arylene, C5-C 14 heteroarylene, bivalent C4-C1 0 heterocycle, each of which can be optionally substituted; or Ri and R2' together form a 4-to 6-membered heterocylic ring comprising the oxygen atom adjacent to Ri and the nitrogen atom adjacent to R2'; provided that the compound has at least one prodrug-forming moiety selected from the group consisting of the first and the second prodrug-forming moieties.

As used herein, the term "amide linkage" comprises a structure represented by the formula:

, wherein 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. As used herein, the term "carbamate linkage" comprises a structure mula:

, wherein 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.

As used herein, the term "phosphoramidate linkage" comprises a structure

in 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.

As used herein, the term "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 R4) m -Q-Z, aryl, substituted aryl, alkylamine, substituted alkylamine, heteroaryl, substituted heteroaryl, and

In some embodiments, X is -CH 2 -, and n is i .

In other embodiments, X is -0-. In some embodiments, the prodrug compound has both the first prodrug-forming moiety and the second prodrug-forming moiety. In some embodiments, the glutamine analog is a glutamine antagonist, i.e., the prodrug is a prodrug of a glutamine analog that antagonizes a glutamine pathway. Exemplary glutamine antagonists include, without limitation, 6-diazo-5-oxo- norleucine (DON), and aza-serine, and 5-diazo-4-oxo-L-norvaline (L-DONV).

In some embodiments, 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).

In some embodiments, Ri of formula (I) comprises a residue PROi of the prodrug-forming moiety, which, together with a basic moiety and the terminal hydroxyl group forms a salt.

In some embodiments, Ri of formula (I) comprises a residue PROi of the prodrug-forming moiety, which, together with an alkyl group and the oxygen of an adjoining hydroxyl group forms an ester.

In some embodiments, Ri of formula (I) comprises a residue PROi 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.

In some embodiments, Ri 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 subsituents include alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl.

In some embodiments, Ri of formula (I) is not H. In some embodiments, Ri of formula (I) is not H when R 2 and R2' are H. In some embodiments, R2 and R2' of formula (I) are each H when and Ri is not H.

In some embodiments, Ri of formula (I) is selected from the group consisting of a Ci-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(Ci-C8-alkyl)ammonium, tetra(Ci-Cg- alkyl)ammonium, triphenylammonium, tri(hydroxy-Ci-C8-alkyl)ammonium, and tetra(hydroxy-Ci-C8-alkyl)ammonium.

In some embodiments, Ri 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. In some embodiments, Ri of formula (I) is methyl. In some embodiments, Ri of formula (I) is ethyl. In some embodiments, Ri of formula (I) is isopropyl.

In some embodiments, R2 of formula (I) comprises a residue PRO2 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.

In some embodiments, R2 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.

In some aspects, a prodrug of a glutamine antagonist, or a pharmaceutically acceptable salt or ester a (I):

(i);

wherein: X is selected from the group consisting of a bond, -0-, and -(CH 2 ) n - wherein n is an integer selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, and 8; Ri is selected from the group consisting of H and a first prodrug-forming moiety capable of forming a salt or an ester; and R2 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 R2; R 2 ' is selected from the group consisting of H, C1-C6 alkyl, substituted C1-C6 alkyl, or R 2 and R2' together form a ring structure comprising -C(=0)-G-C(=0)-, wherein G is selected from the group consisting of Ci-Cg alkylene, Ci-Cg heteroalkylene, Cs-Cg

cycloalkylene, C6-C12 arylene, C5-C 14 heteroarylene, bivalent C4-C1 0 heterocycle, each of which can be optionally substituted; or Ri and R2' together form a 4-to 6-membered heterocylic ring comprising the oxygen atom adjacent to Ri and the nitrogen atom adjacent to R2'; provided that the compound has at least one prodrug-forming moiety selected from the group consisting of the first and the second prodrug-forming moieties.

As used herein, the term "amide linkage" comprises a structure represented by the formula:

wherein 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.

As used herein, the term "carbamate linkage" comprises a structure

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.

As used herein, the term "phosphoramidate linkage" comprises a structure represented by the formula: wherein 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.

As used herein, the term "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

In some embodiments, X is -CH 2 -, and n is i .

In other embodiments, X is -0-. In some embodiments, the prodrug compound has both the first prodrug-forming moiety and the second prodrug-forming moiety. In some embodiments, the glutamine analog is a glutamine antagonist, i.e., the prodrug is a prodrug of a glutamine analog that antagonizes a glutamine pathway. Exemplary glutamine antagonists include, without limitation, 6-diazo-5-oxo- norleucine (DON), and aza-serine, and 5-diazo-4-oxo-L-norvaline (L-DONV).

In some embodiments, 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).

In some embodiments, Ri of formula (I) comprises a residue PROi of the prodrug-forming moiety, which, together with a basic moiety and the terminal hydroxyl group forms a salt.

In some embodiments, Ri of formula (I) comprises a residue PROi of the prodrug-forming moiety, which, together with an alkyl group and the oxygen of an adjoining hydroxyl group forms an ester.

In some embodiments, Ri of formula (I) comprises a residue PROi of the prodrug-forming moiety, which, together with an alkyl group and the nitrogen adjoining the R2' group, forms an azlactone or an oxazolidone.

In some embodiments, Ri 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 subsituents include alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl.

In some embodiments, Ri of formula (I) is not H. In some embodiments, Ri of formula (I) is not H when R 2 and R2' are H. In some embodiments, R2 and R2' of formula (I) are each H when and Ri is not H.

In some embodiments, Ri of formula (I) is selected from the group consisting of a Ci-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(Ci-C8-alkyl)ammonium, tetra(Ci-Cg- alkyl)ammonium, triphenylammonium, tri(hydroxy-Ci-C8-alkyl)ammonium, and tetra(hydroxy-Ci-C8-alkyl)ammonium.

In some embodiments, Ri 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. In some embodiments, Ri of formula (I) is methyl. In some embodiments, Ri of formula (I) is ethyl. In some embodiments, Ri of formula (I) is isopropyl.

In some embodiments, R2 of formula (I) comprises a residue PRO2 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.

In some embodiments, R2 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.

In some embodiments, R2 of formula (I) is selected from the group consisting of H, alkyl, -C(=0)-Ar, -C(=0)-Y-(CR 3 R4) m -Ar, -

0-R 8 , -C(=0)-0-(CR 3 R4) m -0-C(=0)-Rio,-C(=0)-0-R 9 , -C(=0)-Y-(CR 3 R4) m -Ar-0- C(=0)-Ar, and wherein: Y is -O- or a bond; m is an integer selected from the group consisting of 0, 1, 2, 3, 4, 5, 6, 7, and 8; each n and 0 is an integer from 0 to 2 provided that the sum of n and 0 is 2; R 3 and R4 is independently H, C1-C6 alkyl or substituted C1-C6 alkyl, aryl or substituted aryl, -

, alkyl, -

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 R4) m -Z, -(CR 3 R4), Q-Z, wherein Q is a monosaccharide, aryl, substituted aryl, heteroaryl, substituted

heteroaryl, and wherein Z is

or wherein R 7 together with the oxygen atom to which it is attached forms a purine or pyrimidine nucleoside; 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, - aryl, substituted aryl, heteroaryl, substituted heteroaryl, and

, wherein Ri and X are as defined above, provided that Ri 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 R3, R4, R5, and/or R 6 can be made to the 5 or 6 membered rings of Z.

Structures of representative DON prodrugs are provided in Table 1.

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 . The first step in glycolysis is the phosphorylation of glucose by hexokinase to form glucose 6-phosphate.

In some embodiments, 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. In the context of decreasing 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). As used herein, the term "glycolytic metabolic activity" refers to the chemical reactions and enzymes involving the glycolysis pathway.

In some embodiments, 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, phosphogly cerate kinase, phosphogly cerate 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. It should be recognized that a compound that is a useful metabolic reprogramming agent may have two or more of these characteristics.

In some embodiments, at least one metabolic reprogramming agent interferes or inhibits the expression and/or activity of hexokinase. Examples of inhibitors of hexokinase include, but are not limited to, 2-deoxy glucose (2-DG), 3-bromopyruvate (3-BrPA), lonidamine (LND), sodium fluoride, and potassium fluoride. In some embodiments, 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.

In some embodiments, 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.

Certain methods, compositions, and metabolic reprogramming agents contemplated herein decrease fatty acid synthesis and/or increase fatty acid oxidation in cells. In the context of decreasing fatty acid synthesis and/or increasing 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).

In some embodiments, 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 β-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. It should be recognized that a compound that is a useful metabolic reprogramming agent may have two or more of these characteristics. In some embodiments, at least one metabolic reprogramming agent is an activator of 5' AMP- activated protein kinase (AMPK) activity.

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-imidazolecarboxarnide 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.

In some embodiments, at least one metabolic reprogramming agent is metformin.

It should be appreciated that 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.

The term "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. Those skilled in the art will appreciate that the method of modulating 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.). Those skilled in the art will readily appreciate 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.). For example, 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. The metabolic reprogramming agents, cytotoxic agents, immunotherapeutic agents, immunosuppressant agents, radiotherapeutic agents, anti-inflammatory agents, and neuroprotective agents described herein can be any type of agent. Exemplary types of 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.

As used herein, the term "small molecule" can refer to agents that are "natural product-like," however, the term "small molecule" is not limited to "natural productlike" 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.

As used herein, an "RNA interference molecule" refers to an agent which interferes with or inhibits expression of a target gene or genomic sequence by RNA interference (RNAi). Such 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).

The term "polynucleotide" is used herein interchangeably with "nucleic acid" to indicate a polymer of nucleosides. Typically 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 deoxy cytidine) joined by phosphodiester bonds. However 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. Where this application refers to a polynucleotide it is understood that both DNA, RNA, and in each case both single- and double-stranded forms (and complements of each single-stranded molecule) are provided.

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 US 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. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.

The terms "polypeptide" as used herein refers to a polymer of amino acids. The terms "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. A 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.

The term "identity" as used herein 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, e.g. , over the length of the sequence of interest, 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. When computing the number of identical residues needed to achieve a particular percent identity, 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. The algorithm of Karlin and Altschul (Karlin and Altschul, Proc. Natl. Acad. Sci. USA 87:22264-2268, 1990) modified as in 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). To obtain gapped alignments for comparison purposes, Gapped BLAST is utilized as described in Altschul et al. (Altschul, et al. Nucleic Acids Res. 25: 3389-3402, 1997). When utilizing BLAST and Gapped BLAST programs, 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. In a specific embodiment, percent identity is calculated using BLAST2 with default parameters as provided by the NCBI.

Generally, 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). 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. Examples of 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, immunotherapeutic agents, immunosuppressant agents, anti-inflammatory agents, neuroprotective agents, neuroregenerative agents, neurotrophic factors, radiotherapeutic agents, and stem and progenitor cells used to replace and/or repair endogenous populations of abnormal, harmful, or unhealthy cells.

As used herein, the term "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. Examples of 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 adoptive T cell therapy including chimeric antigen receptor T cells (CAR T cells).

As used herein, "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. Examples of 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. These include, without limitation, CCI-779, ABT578, SAR543, rapamycin and derivatives or analogs thereof, including 40-0(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 Rapamycin 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.

As used herein, "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. In some embodiments, antiinflammatory 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 dipropionate, diclofenac potassium, diclofenac sodium, diflorasone diacetate, diflumidone sodium, diflunisal,

difluprednate, diftalone, dimethyl sulfoxide, drocinonide, endrysone, enlimomab, enolicam sodium, epirizole, etodolac, etofenamate, felbinac, fenamole, fenbufen, fenclofenac, fenclorac, fendosal, fenpipalone, fentiazac, flazalone, fluazacort, flufenamic acid, flumizole, flunisolide acetate, flunixin, flunixin meglumine, fluocortin butyl, fluorometholone acetate, fluquazone, flurbiprofen, fluretofen, fluticasone propionate, furaprofen, furobufen, halcinonide, halobetasol propionate, halopredone acetate, ibufenac, ibuprofen, ibuprofen aluminum, ibuprofen piconol, ilonidap, indomethacin, indomethacin sodium, indoprofen, indoxole, intrazole, isoflupredone acetate, isoxepac, isoxicam, ketoprofen, lofemizole hydrochloride, lomoxicam, loteprednol etabonate, meclofenamate sodium, meclofenamic acid, meclorisone dibutyrate, mefenamic acid, mesalamine, meseclazone,

methylprednisolone suleptanate, momiflumate, nabumetone, naproxen, naproxen sodium, naproxol, nimazone, olsalazine sodium, orgotein, orpanoxin, oxaprozin, oxyphenbutazone, paranyline hydrochloride, pentosan polysulfate sodium, phenbutazone sodium gly cerate, pirfenidone, piroxicam, piroxicam cinnamate, piroxicam olamine, pirprofen, prednazate, prifelone, prodolic acid, proquazone, proxazole, proxazole citrate, rimexolone, romazarit, salcolex, salnacedin, salsalate, sanguinarium chloride, seclazone, sermetacin, sudoxicam, sulindac, suprofen, talmetacin, talniflumate, talosalate, tebufelone, tenidap, tenidap sodium, tenoxicam, tesicam, tesimide, tetrydamine, tiopinac, tixocortol pivalate, tolmetin, tolmetin sodium, triclonide, triflumidate, zidometacin, zomepirac sodium, aspirin

(acetylsalicylic acid), salicylic acid, corticosteroids, glucocorticoids, tacrolimus, pimecorlimus, prodrugs thereof, co-drugs thereof, and combinations thereof. The antiinflammatory agent may also be a biological inhibitor of proinflammatory signaling molecules including antibodies to such biological inflammatory signaling molecules.

Exemplary 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. 13:466-80 (2010)); serotonin receptor agonists; continuous- release levodopa (Sinemet CR.RTM., MSD, Israel); continuous duodenal levodopa administration (Duodopa.RTM., Abbott, UK); catechol-O-methyltransferase (COMT) inhibitors (e.g., Stalevo.RTM., Novartis Pharma, USA; entacapone (Comtan.RTM., Novartis Pharma, USA)); tolcapone; coenzyme Q10, and/or MAO-B inhibitors (e.g., Selegiline or Rasagiline). Additional neuroprotective agents are described in, e.g., Hart et al, Mov. Disord. 24: 647-54 (2009).

As used herein, 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 (β), and gamma (γ) 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 yrays 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. In case of internal radiation therapy, 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. A unit dose of radiation is generally measured in gray (Gy). 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. In the latter case, the balloon has been specially configured to prevent leakage of the radioisotopic material from the balloon into the body lumen or blood stream. Still further, 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 α, β and γ. Usually, a and β 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 γ-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. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the target cell. In some embodiments, the radiotherapeutic agent is selected from the group consisting of 47 Sc, 67 Cu, 90 Y, 109 Pd, 123 I, 125 I, m 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, i n Ag, 119 Sb, 121 Sn, 131 Cs, 143 Pr,

described in U.S. Pat. No. 8,946,168, the entirety of which is incorporated herein by reference.

In some contexts, an agent described herein can be administered with an antigen (e.g., to induce an immune response). In some embodiments, 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) 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. Many appropriate 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.), intratracheal, or intrathecal 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. In some contexts, the imaging agent can be used for detecting and/or monitoring tumors or sites of metastasis in a subject. For example, an agent (e.g., a metabolic reprogramming agent) can be administered in vivo and monitored using an appropriate label.

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. For example, 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-I l l, 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.

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-I l l, 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-I l l, Platinum- 197, Palladium- 109, Copper-67, Phosphorus-32, Phosphorus-33, Yttrium-90, Scandium-47, Samarium-153, Lutetium-177, Rhodium- 105, Praseodymium- 142, Praseodymium- 143, Terbium-161, Holmium-166, Gold- 199, Cobalt-57, Cobalt-58, Chromium-51, Iron-59, Selenium-75, Thallium-201, and Ytterbium- 169. The most preferred radioisotope for use in the current invention is Technetium-99m. Preferably 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-I l l, 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.

III. USES OF METABOLIC REPROGRAMMING AGENTS

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. Accordingly, in an aspect 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.

In some embodiments, 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.

In an aspect, 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 arthritis in a subject in need thereof. In an aspect, 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 lupus in a subject in need thereof.

In an aspect, 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 asthma in a subject in need thereof.

In an aspect, 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 acute respiratory distress syndrome (ARDS) in a subj ect in need thereof.

In an aspect, 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 pulmonary fibrosis in a subject in need thereof.

In an aspect, the presently disclosed subject matter involves the use of an effective amount of at least one metabolic reprogramming agent that decreases glutamine metabolism to prevent or delay allograft rejection during cell, tissue, or organ transplantation in a subject in need thereof.

In an aspect, 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 or prevent graft versus host disease in a subject in need thereof.

In an aspect, 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 multiple sclerosis in a subject in need thereof.

In an aspect, 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 neuromyelitis optica in a subject in need thereof.

In an aspect, 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 cerebral malaria in a subject in need thereof.

In an aspect, 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 a pathology due to CNS inflammation involving an infection in a subject in need thereof. In an aspect, 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 a pathology due to CNS inflammation not involving an infection in a subject in need thereof.

In an aspect, 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 a Alzheimer's disease in a subject in need thereof.

In an aspect, 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 a Parkinson's disease in a subject in need thereof.

In an aspect, 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 amyotrophic lateral sclerosis (ALS) in a subject in need thereof.

In an aspect, 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 pneumonitis in a subject in need thereof.

In some embodiments, 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. IV. PHARMACEUTICAL COMPOSITIONS COMPRISING METABOLIC REPROGRAMMING AGENTS

The presently disclosed subject matter also contemplates pharmaceutical compositions comprising one or more metabolic reprogramming agents for the treatment of certain conditions, diseases, and/or disorders involving metabolically reprogrammed cells. In some embodiments, 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.

Accordingly, in an aspect the presently disclosed subject matter provides 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.

In some embodiments, the metabolic reprogramming composition comprises one or more additional therapeutic agents described herein. Generally, the presently disclosed compositions (e.g., comprising at least one metabolic reprogramming agent) 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, or intraocular injections, intracisternally, topically, as by powders, ointments or drops (including eyedrops), including buccally and sublingually, trans dermally, through an inhalation spray, or other modes of delivery known in the art.

The phrases "systemic administration", "administered systemically",

"peripheral administration" and "administered peripherally" as used herein mean 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.

The phrases "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 intrasternal injection and infusion.

The presently disclosed pharmaceutical 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.

In some embodiments, the presently disclosed pharmaceutical compositions can be administered by rechargeable or biodegradable devices. For example, 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. Patent No. 3,773,919; EP 58,481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al, Biopolymers 22:547, 1983), poly (2-hydroxyethyl-methacrylate) (Langer et al. (1981) J. Biomed. Mater. Res. 15: 167; Langer (1982), Chem. Tech. 12:98), ethylene vinyl acetate (Langer et al. (1981) J. Biomed. Mater. Res. 15: 167), or poly-D-(-)-3- hydroxybutyric acid (EP 133,988A). 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. Patent Nos. 4,485,045 and 4,544,545; and EP 102,324 A). Ordinarily, 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, predetermined target site.

In another embodiment, 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). Some effects of PEGylation 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.al. (1992) J. Immunol. Meth. 152: 177-190); (f) improved bioavailability via reduced losses at subcutaneous injection sites; (g) reduced toxicity has been observed; for agents where toxicity is related to peak plasma level, a flatter pharmacokinetic profile achieved by sub-cutaneous administration of PEGylated protein is advantageous; proteins that elicit an immune response which has toxicity consequences may also benefit as a result of PEGylation; and (h) improved thermal and mechanical stability of the PEGylated molecule.

Pharmaceutical compositions for parenteral administration include aqueous solutions of compositions comprising at least one metabolic reprogramming agent. For injection, 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. Additionally, suspensions of compositions include fatty oils, such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Optionally, 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.

For nasal or transmucosal administration generally, penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.

Additional ingredients 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. For example, 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.

Regardless of the route of administration selected, the presently disclosed 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.

In general, 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. As will be appreciated by those of ordinary skill in this art, 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. As used herein, 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. In one embodiment of the presently disclosed subject matter, the active agents are combined and administered in a single dosage form. In another embodiment, 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.

Further, the presently disclosed 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. Advantageously, such 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. Therefore, 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.

When administered sequentially, the agents 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 subj ect 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.

When administered in combination, 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.

In some embodiments, when administered in combination, the two or more agents can have a synergistic effect. As used herein, 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:

Q 3 QA + QbQ B = Synergy Index (SI) wherein:

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; and

Qb is the concentration of component B, in a mixture, which produced an end point.

Generally, when the sum of Q 3 /Q A and QI/Q B is greater than one, antagonism is indicated. When the sum is equal to one, additivity is indicated. When the sum is less than one, synergism is demonstrated. The lower the SI, the greater the synergy shown by that particular mixture. Thus, a "synergistic combination" has an activity higher that what can be expected based on the observed activities of the individual components when used alone. Further, 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.

In another aspect, the presently disclosed subj ect 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.

More particularly, the presently disclosed subject matter provides 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.

In therapeutic and/or diagnostic applications, 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).

Use of pharmaceutically acceptable inert carriers to formulate the compounds herein disclosed for the practice of the disclosure into dosages suitable for systemic administration is within the scope of the disclosure. With proper choice of carrier and suitable manufacturing practice, the 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.

For nasal or inhalation delivery, 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.

Pharmaceutical 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.

In addition to the active ingredients, 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. The preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.

The term "instructing" a patient as used herein 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.

The term "promoting" as used herein 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.

V. GeneralDefinitions

Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this presently described subject matter belongs.

While the following terms in relation to compounds of Formula (I) are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter. These definitions are intended to supplement and illustrate, not preclude, the definitions that would be apparent to one of ordinary skill in the art upon review of the present disclosure.

The terms substituted, whether preceded by the term "optionally" or not, and substituent, as used herein, refer 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. When more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the 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).

Where 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 0- is equivalent to -OCH 2 -; -C(=0)0- is equivalent to -OC(=0)-;

-OC(=0)NR- is equivalent to -NRC(=0)0-, and the like.

When the term "independently selected" is used, the substituents being referred to (e.g., R groups, such as groups R 1; R 2 , and the like, or variables, such as

"m" and "n"), can be identical or different. For example, both Ri and R 2 can be substituted alkyls, or Ri can be hydrogen and R 2 can be a substituted alkyl, and the like.

The terms "a," "an," or "a(n)," when used in reference to a group of substituents herein, mean at least one. For example, where 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. Moreover, where a moiety is substituted with an R substituent, 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.

A named "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. For the purposes of illustration, certain representative "R" groups as set forth above are defined below.

Descriptions of compounds of the present disclosure are limited by principles of chemical bonding known to those skilled in the art. Accordingly, where a group may be substituted by one or more of a number of substituents, such substitutions are selected so as to comply with principles of chemical bonding and to give compounds which are not inherently unstable and/or would be known to one of ordinary skill in the art as likely to be unstable under ambient conditions, such as aqueous, neutral, and several known physiological conditions. For example, a heterocycloalkyl or heteroaryl is attached to the remainder of the molecule via a ring heteroatom in compliance with principles of chemical bonding known to those skilled in the art thereby avoiding inherently unstable compounds.

Unless otherwise explicitly defined, a "substituent group," as used herein, includes a functional group selected from one or more of the following moieties, which are defined herein:

The term hydrocarbon, as used herein, 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, poly cyclic, or heterocyclic. Illustrative hydrocarbons are further defined herein below and include, for example, methyl, ethyl, ^-propyl, isopropyl, cyclopropyl, allyl, vinyl, w-butyl, fert-butyl, ethynyl, cyclohexyl, and the like.

The term "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., Ci-Cio means one to ten carbons, including 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10 carbons). In particular embodiments, the term "alkyl" refers to Ci-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.

Representative saturated hydrocarbon groups include, but are not limited to, methyl, ethyl, ^-propyl, isopropyl, w-butyl, isobutyl, seobutyl, fert-butyl, w-pentyl, seopentyl, isopentyl, neopentyl, w-hexyl, seohexyl, w-heptyl, w-octyl, «-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. In certain embodiments, "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. The term "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. 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.

Thus, as used herein, the term "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.

The term "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 quatemized. 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 -0-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(0)-CH 3 ,

-CH 2 -CH 2 -S(0) 2 -CH 3 , -CH=CH-0-CH 3 , -Si(CH 3 ) 3 , -CH 2 -CH=N-OCH 3 ,

-CH=CH-N(CH 3 )- CH 3 , 0-CH 3 , -0-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 -0-Si(CH 3 ) 3 .

As described above, heteroalkyl groups, as used herein, include those groups that are attached to the remainder of the molecule through a heteroatom, such as -C(0)NR', -NR'R", -OR', -SR, -S(0)R, and/or -S(0 2 )R' . Where "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. 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.

The term "cycloalkylalkyl," as used herein, refers to a cycloalkyl group as defined hereinabove, which is attached to the parent molecular moiety through an alkyl group, also as defined above. Examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.

The terms "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.

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 quatemized. In certain embodiments, the term heterocylic refers to a non-aromatic 5-, 6-, or 7- membered ring or a poly cyclic 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 quatemized, 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.

The terms "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. Examples of heterocycloalkyl 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. The terms

"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. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2- propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(l,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." More particularly, the term "alkenyl" as used herein refers to a monovalent group derived from a C 1-2 o 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-l-yl, pentenyl, hexenyl, octenyl, allenyl, and butadienyl.

The term "cycloalkenyl" as used herein 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.

The term "alkynyl" as used herein refers to a monovalent group derived from a straight or branched C 1-2 o hydrocarbon of a designed number of carbon atoms containing at least one carbon-carbon triple bond. Examples of "alkynyl" include ethynyl, 2-propynyl (propargyl), 1-propynyl, pentynyl, hexynyl, and heptynyl groups, and the like.

The term "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, 1 1, 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. Exemplary alkylene groups include methylene (-CH 2 -); ethylene (-CH 2 -CH 2 -); propylene (-(CH 2 ) 3 -);

cyclohexylene (-C 6 Hi 0 -); -CH=CH-CH=CH-; -CH=CH-CH 2 -; -CH 2 CH 2 CH 2 CH 2 -, -CH 2 CH=CHCH 2 -, -CH2CSCCH2-, -CH 2 CH2CH(CH2CH 2 CH3)CH2-,

-(CH2)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, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, or 20, and R is hydrogen or lower alkyl; methylenedioxyl (-0-CH 2 -0-); and

ethylenedioxyl (-0-(CH 2 ) 2 -0-). 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.

The term "heteroalkylene" by itself or as part of another substituent means a divalent group derived from heteroalkyl, as exemplified, but not limited by,

-CH2-CH2-S-CH2-CH2- and -CH2-S-CH2-CH2-NH-CH2-. For heteroalkylene groups, heteroatoms also can occupy either or both of the chain termini (e.g., alkyleneoxo, alkylenedioxo, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. For example, the formula -C(0)OR'- represents both -C(0)OR'- and -R'OC(O)-.

The term "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. The term "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 quatemized. 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- isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl, and 6-quinolyl. Substituents for each of above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below. The terms "arylene" and "heteroarylene" refer to the divalent forms of aryl and heteroaryl, respectively.

For brevity, the term "aryl" when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, the terms "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-(l-naphthyloxy)propyl, and the like). However, the term "haloaryl," as used herein is meant to cover only aryls substituted with one or more halogens.

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.

Further, a structure represented generally by the formula:

as used herein refers to 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. Each R group, if more than one, is substituted on an available carbon of the ring structure rather than on another R group. For example, the structure above where n is 0 to 2 would comprise compound groups including, but not limited to:

and the like.

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.

The symbol ( ,ΛΛΛΛΛΛΛ Λ ' ) denotes the point of attachment of a moiety to the remainder of the molecule.

When a named atom of an aromatic ring or a heterocyclic aromatic ring is defined as being "absent," the named atom is replaced by a direct bond.

Each of above terms (e.g. , "alkyl," "heteroalkyl," "cycloalkyl, and

"heterocycloalkyl", "aryl," "heteroaryl," "phosphonate," and "sulfonate" as well as their divalent derivatives) are meant to include both substituted and unsubstituted forms of the indicated group. Optional substituents for each type of group are provided below.

Substituents for alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl monovalent and divalent derivative groups (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be one or more of a variety of groups selected from, but not limited to: -OR', =0, =NR', =N-OR', -NR'R", -SR', -halogen, -SiR'R"R'", -OC(0)R', -C(0)R', -C0 2 R',-C(0)NR'R", -OC(0)NR'R", - NR"C(0)R', -NR'-C(0)NR"R'", -NR"C(0)OR', -NR-C(NR'R")=NR'", -S(0)R', - S(0) 2 R', -S(0) 2 NR'R", -NRSO 2 R', -CN and -N0 2 in a number ranging from zero to (2m' +1), where m' is the total number of carbon atoms in such groups. 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. As used herein, an "alkoxy" group is an alkyl attached to the remainder of the molecule through a divalent oxygen. When a compound of the disclosure includes more than one R group, for example, 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. When 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. For example, -NR'R" is meant to include, but not be limited to, 1- pyrrolidinyl and 4- morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term "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(0)CH 3 , -C(0)CF 3 , -C(0)CH 2 OCH 3 , and the like).

Similar to the substituents described for alkyl groups above, exemplary substituents for aryl and heteroaryl groups (as well as their divalent derivatives) are varied and are selected from, for example: halogen, -OR', -NR'R", -SR',

-SiR'R"R"', -OC(0)R', -C(0)R', -C0 2 R', -C(0)NR'R", -OC(0)NR'R", - NR"C(0)R', -NR'-C(0)NR"R"', -NR"C(0)OR', -NR-C(NR'R"R"')=NR"", -NR-C(NR'R")=NR'" -S(0)R', -S(0) 2 R', -S(0) 2 NR'R", -NRS0 2 R', -CN and -N0 2 , -R', -N 3 , -CH(Ph) 2 , fluoro(Ci-C4)alkoxo, and fluoro(Ci-C4)alkyl, in a number ranging from zero to the total number of open valences on aromatic ring system; and where R', R", R'" and R"" may be independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl and substituted or unsubstituted heteroaryl. When a compound of the disclosure includes more than one R group, for example, 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.

Two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally form a ring of the formula -T-C(0)-(CRR') q -U-, wherein T and U are independently -NR-, -0-, -CRR'- or a single bond, and q is an integer of from 0 to 3. Alternatively, 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'-, -0-, -NR-, -S-, -S(O)-, -S(0) 2 -, -S(0) 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. Alternatively, 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 -0-, -NR'-, -S-, -S(O)-, -S(0) 2 -, or -S(0) 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.

As used herein, the term "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(=0)-, wherein R is an alkyl, alkenyl, alkynyl, aryl, carbocylic, heterocyclic, or aromatic heterocyclic group as defined herein). As such, the term "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(=0)NR', esters, -RC(=0)OR', ketones, -RC(=0)R', and aldehydes, -RC(=0)H.

The terms "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 1-2 o inclusive, linear, branched, or cyclic, saturated or unsaturated oxo-hydrocarbon chains, including, for example, methoxyl, ethoxyl, propoxyl, isopropoxyl, w-butoxyl, seobutoxyl, fert-butoxyl, and ^-pentoxyl, neopentoxyl, w-hexoxyl, and the like.

The term "alkoxy alkyl" as used herein 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. The term "aryloxyl" as used herein can refer to phenyloxyl or hexyloxyl, and alkyl, substituted alkyl, halo, or alkoxyl substituted phenyloxyl or hexyloxyl.

"Aralkyl" 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.

"Aralkyloxyl" refers to an aralkyl-O- group wherein the aralkyl group is as previously described. An exemplary aralkyloxyl group is benzyloxyl, i.e.,

C6H5-CH2-O-. An aralkyloxyl group can optionally be substituted.

"Alkoxycarbonyl" refers to an alkyl-0-C(=0)- group. Exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, butyloxycarbonyl, and fert-butyloxycarbonyl.

"Aryloxycarbonyl" refers to an aryl-0-C(=0)- group. Exemplary

aryloxycarbonyl groups include phenoxy- and naphthoxy-carbonyl.

"Aralkoxycarbonyl" refers to an aralkyl-0-C(=0)- group. An exemplary aralkoxycarbonyl group is benzyloxycarbonyl.

"Carbamoyl" refers to an amide group of the formula -C(=0)NH 2 .

"Alkylcarbamoyl" refers to a R'RN-C(=0)- 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(=0)- group wherein each of R and R' is independently alkyl and/or substituted alkyl as previously described. The term carbonyldioxyl, as used herein, refers to a carbonate group of the formula -0-C(=0)-OR.

"Acyloxyl" refers to an acyl-O- group wherein acyl is as previously described.

The term "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. For example, the terms

"acylamino" and "alkylamino" refer to specific N-substituted organic radicals with acyl and alkyl substituent groups respectively.

An "aminoalkyl" as used herein 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. The term 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. The term 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.

The terms 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. Examples of thioalkoxyl moieties include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, w-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. The term "carbonyl" refers to the -C(=0)- group, and can include an aldehyde group represented by the general formula R-C(=0)H.

The term "carboxyl" refers to the -COOH group. Such groups also are referred to herein as a "carboxylic acid" moiety.

The terms "halo," "halide," or "halogen" as used herein refer to fluoro, chloro, bromo, and iodo groups. Additionally, terms, such as "haloalkyl," are meant to include monohaloalkyl and polyhaloalkyl. For example, the term "halo(Ci-C4)alkyl" is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4- chlorobutyl, 3-bromopropyl, and the like.

The term "hy droxyl" refers to the -OH group.

The term "hydroxyalkyl" refers to an alkyl group substituted with an -OH group.

The term "mercapto" refers to the -SH group.

The term "oxo" as used herein means an oxygen atom that is double bonded to a carbon atom or to another element.

The term "nitro" refers to the -NO2 group.

The term "thio" refers to a compound described previously herein wherein a carbon or oxygen atom is replaced by a sulfur atom.

The term "sulfate" refers to the -SO4 group.

The term thiohy droxyl or thiol, as used herein, refers to a group of the formula

-SH.

More particularly, the term "sulfide" refers to compound having a group of the formula -SR.

The term "sulfone" refers to compound having a sulfonyl group -S(02)R. The term "sulfoxide" refers to a compound having a sulfinyl group -S(0)R

The term ureido refers to a urea group of the formula -NH— CO— NH 2 .

Throughout the specification and claims, a given chemical formula or name shall encompass all tautomers, congeners, and optical- and stereoisomers, as well as racemic mixtures where such isomers and mixtures exist.

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. When 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.

Unless otherwise stated, 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.

It will be apparent to one skilled in the art that certain compounds of this disclosure may exist in tautomeric forms, all such tautomeric forms of the compounds being within the scope of the disclosure. The term "tautomer," as used herein, refers to one of two or more structural isomers which exist in equilibrium and which are readily converted from one isomeric form to another.

Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures with the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 1 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. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 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. Also included are base addition salts, such as sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present disclosure contain relatively basic functionalities, 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. Examples of 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. Also included are salts of amino acids, such as arginate and the like, and 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.

In addition to salt forms, 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. Additionally, 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.

Following long-standing patent law convention, the terms "a," "an," and "the" refer to "one or more" when used in this application, including the claims. Thus, for example, reference to "a subject" includes a plurality of subjects, unless the context clearly is to the contrary (e.g., a plurality of subjects), and so forth.

Throughout this specification and the claims, the terms "comprise,"

"comprises," and "comprising" are used in a non-exclusive sense, except where the context requires otherwise. Likewise, the term "include" and its grammatical variants are intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that can be substituted or added to the listed items.

For the purposes of this specification and appended claims, unless otherwise indicated, all numbers expressing amounts, sizes, dimensions, proportions, shapes, formulations, parameters, percentages, quantities, characteristics, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term "about" even though the term "about" may not expressly appear with the value, amount or range. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are not and need not be exact, but may be approximate and/or larger or smaller as desired, reflecting tolerances, conversion factors, rounding off, measurement error and the like, and other factors known to those of skill in the art depending on the desired properties sought to be obtained by the presently disclosed subject matter. For example, 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.

Further, the term "about" when used in connection with one or more numbers or numerical ranges, should be understood to refer to all such numbers, including all numbers in a range and modifies that range by extending the boundaries above and below the numerical values set forth. The recitation of numerical ranges by endpoints includes all numbers, e.g., whole integers, including fractions thereof, subsumed within that range (for example, the recitation of 1 to 5 includes 1 , 2, 3, 4, and 5, as well as fractions thereof, e.g., 1.5, 2.25, 3.75, 4.1, and the like) and any range within that range.

EXAMPLES

The following Examples have been included to provide guidance to one of ordinary skill in the art for practicing representative embodiments of the presently disclosed subject matter. In light of the present disclosure and the general level of skill in the art, those of skill can appreciate that the following Examples are intended to be exemplary only and that numerous changes, modifications, and alterations can be employed without departing from the scope of the presently disclosed subject matter. The synthetic descriptions and specific examples that follow are only intended for the purposes of illustration, and are not to be construed as limiting in any manner to make compounds of the disclosure by other methods. EXAMPLE 1

Treatment of Immune Disorders

The presently disclosed glutamine analogues were found to suppress antigen- specific effector CD4 + T cells proliferation and function (FIG. 1C, FIG. ID, and FIG. IE) and antigen-specific CD8 + T cells responses in vivo (FIG. 2A, FIG. 2B, and FIG. 2C). The addition of DON to the metabolic reprogramming therapy resulted in very potent inhibition of both CD4 + T cell effector function and CD8 + T cell effector function.

To determine if this suppression of T cells by the glutamine analogues could be used to reduce transplant rejections, skin from Balb/c mice was transplanted onto B6 mice and the mice were treated with the metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + Metformin + DON) or DON alone (FIG. 3). The results showed that DON prevents transplant rejection and prolongs skin graft survival. Further, when hearts from Balb/c mice were transplanted into B6 mice, it was found that DON is able to prevent heart transplant rejection (FIG. 4). DON also increased the relative frequency of regulatory T cells (Tregs) in vivo (FIG. 5).

To further identify the effect of DON on the immune system, different mouse models showed that the metabolic reprogramming therapy with at least three metabolic reprogramming agents targeted asthmatic cells (FIG. 6), and that DON alone treated arthritis (FIG. 7), and treated pneumonitis (FIG. 8).

EXAMPLE 2

Regulation of Immune Metabolism as the Sole Means of Preventing Allograft

Rejection

Summary

Upon antigen recognition and co-stimulation, T lymphocytes up-regulate the metabolic machinery necessary to proliferate and sustain effector function. Hallmarks of this response include a switch to aerobic glycolysis and an increased demand for glutamine. This metabolic reprogramming in T cells is a critical regulator of T cell activation and differentiation, not just a consequence of antigen recognition.

Furthermore, it is clear that while such metabolic reprogramming is important for the differentiation and function of T effector cells, the differentiation of regulatory T cells employs different metabolic reprogramming. Based on these observations, it was hypothesized that inhibition of glycolysis and glutamine metabolism might represent a novel means of preventing graft rejection by inhibiting effector generation and function while promoting regulatory T cell generation. Along these lines, an anti- rejection regimen was devised employing a glycolytic inhibitor, 2-Deoxy glucose (2- DG), an anti-Type II diabetes drug (metformin) and an inhibitor of glutamine metabolism, 6-Diazo-5-oxo-L-norleucine (DON). Using this metabolic

reprogramming therapy with at least three metabolic reprogramming agents, graft rejection was prevented or delayed in fully mismatched skin and heart allograft transplantation models. The data suggest that metabolic reprogramming metabolic reprogramming therapy represents a novel and selective means of inhibiting T cell effector function while preserving mechanisms of immune regulation.

Significance

Transplantation remains the treatment of choice for patients with end-stage organ disease. However, since current mainstream anti-rejection regimens result in multiple adverse effects, new strategies to promote graft survival are of great clinical interest. Metabolic signaling pathways have emerged as playing critical roles in dictating the outcome of T cell responses. Quiescent T cells primarily use oxidative phosphorylation, whereas activated T cells must undergo metabolic reprogramming, which promotes glycolysis and glutamine oxidation to meet the biosynthetic and energy demands. It is demonstrated herein that selective targeting of T cell proliferation and function by blocking glycolysis and glutamine metabolic pathways can inhibit effector T cells response while preserving the mechanisms of immune regulation. Such a regimen can inhibit acute rejection and promote graft survival. Materials and Methods

Mice: Mice were kept in accordance with guidelines of the Johns Hopkins University Institutional Animal Care and Use Committee. 5C.C7 mice (RAG2 " _ CD4 + TCR-transgenic mice which recognize Pigeon cytochrome c (PCC)) were from Taconic Farms. OVA-specific OT-I and OT-II TCR transgenic mice from The Jackson Laboratory were bred to Thy 1.1 + backgrounds. C57BL/6 (Thy 1.2 + , H-2 b ) and Balb/c (H-2 d ) mice were obtained from The Jackson Laboratory.

Antibodies and reagents: The following antibodies for flow cytometry were from BD Biosciences: anti-CD4 (RM4-5), anti-CD8 (Ly-3), anti-IFN-γ (XMG1.2), and anti-Thyl. l (OX-7). Anti-Foxp3 (FJK-16s) was from eBioscience. Class I OVA peptide and class II OVA peptide were obtained from AnaSpec. PCC peptide 81-104 was synthesized at Johns Hopkins University. 2-DG was purchased from Carbosynth. Metformin and DON were purchased from Sigma- Aldrich. For all in vivo

experiments, individual metabolic reprogramming agents were dissolved in PBS and administrated intraperitoneally.

Cell culture: Splenocytes or T cells were cultured in 45% RPMI 1640 and

45% EHAA media supplemented with 10% FBS, penicillin/streptomycin, glutamine and BME. For proliferation studies, T cells were labeled with 5 μΜ eFluor 670 cell proliferation dye (eBioscience) and were stimulated with anti-CD3 (^g/ml). For non-antigen specific stimulations, flat-bottomed plates were coated with anti-CD3 (5 μg/ml) diluted in PBS, and soluble anti-CD28 (2 μg/ml) was added to the cultures. For preparation of pre-activated CD4 + T cells, splenocytes from 5C.C7 mice were stimulated with 5 μΜ PCC peptide in complete medium for 48 hrs. Mouse recombinant IL-2 (1 ng/ml; Peprotech) was then added to the culture. After 5-7 days, live cells were isolated by a Ficoll gradient (GE healthcare). Cell viability was determined by 7-aminoactinomycin D (7-AAD; BD Biosciences) exclusion.

Extracellular flux analysis ; Cells were initially plated to XF assay medium, modified DMEM (Seahorse Bioscience) containing 25 mM glucose, 2 mM L- glutamine, and 1 mM sodium pyruvate, and incubated in a non-C0 2 incubator at 37°C for 30 minutes. OCR and ECAR were measured at 37°C in a XF96 Extracellular Flux Analyzer (Seahorse Bioscience) using manufacturer recommended protocols. After baseline measurements, OCR and ECAR were measured after sequentially adding anti-CD3/CD28 and metabolic reprogramming agents into each well to the indicated final concentrations using the included ports on the XF96 cartridges. All data were collected using the XF Reader software from Seahorse Bioscience.

Adoptive transfers: CD8 + (or CD4 + ) T cells were harvested from OT-I (or OT- II) mice and purified by negative selection with CD8 + (or CD4 + ) MACS cell isolation protocol (Miltenyi Biotec). 1 χ 10 6 purified CD8 + (or CD4 + ) T cells were then injected intravenously into Thyl .2 + C57BL/6 recipients. Vaccinia-OVA (1 χ 10 6 plaque-forming units) was simultaneously administered by injection into the retro- orbital cavernous.

Glutaminase activity analysis: Glutaminase activity measurement was adapted from previously described protocols (Thomas et al. (2013) Biochemical and

Biophysical Research Communications 438(2):243-248). Briefly, T cells were lysed in ice-cold potassium phosphate buffer (45 mM, pH 8.2) containing protease inhibitors (Roche, Complete Protease Inhibitor Cocktail, 1 tablet in

50 ml) and incubated with 3H-glutamine (0.03 μΜ, 0.91 μθί) for 90 min at room temperature. The reactions were carried out in 50 μΐ reaction volumes in a 96-well microplate. At the end of the reaction period, the assay was terminated upon the addition of imidazole buffer (20 mM, pH 7). 96-well spin columns packed with strong anion ion-exchange resin (Bio-Rad, AG® 1-X2 Resin, 200-400 mesh, chloride form) were used to separate the substrate and the reaction product. Unreacted H- glutamine was removed by washing with imidazole buffer. H-glutamate, the reaction product, was then eluted with 0.1 N HC1 and analyzed for radioactivity using Perkin Elmer's TopCount instrument in conjunction with their 96-well LumaPlates.

Flow cytometry, intracellular cytokine staining, andELISA: Flow cytometry data were acquired with a FACS Calibur (BD Biosciences) and were analyzed with FlowJo7.6 software (TreeStar). For intracellular staining, cells were stimulated at 37°C for 4 hrs in the presence of Monensin (GolgiStop; BD Biosciences), phorbol 12- myristate 13- acetate (PMA; Sigma), and ionomycin (Sigma). Cells were surface stained and underwent fixation/permeabilization with either a Cytofix/Cytoperm kit (BD Biosciences) or a Fixation/Permeabilization kit (eBioscience), followed by staining for intracellular cytokines. Gates were determined appropriately through the use of unstimulated control cells. Voltages were determined from unstained controls. IFN-γ concentration in the supernatant of cell cultures was analyzed by ELISA as recommended by the manufacturer (eBioscience).

In vivo cytotoxicity assay: Splenocytes from WT mice were labeled with either 20 μΜ or 2 μΜ of carboxyfluorescein diacetate succinimidyl (CFSE, Invitrogen). The CSFE hi h cells were pulsed with class I OVA peptide (10 μΜ). Equal numbers (5 10 6 cells) of peptide-pulsed CFSE^ 11 cells and impulsed CFSE low cells were coadministered i.v. into syngeneic host mice that had been previously (day 7) immunized with vaccinia-OVA (1 χ 10 6 plaque-forming units). After 10 hrs, splenocytes were isolated from host spleens. The two target populations were distinguished based on the differences in their CFSE intensity by flow cytometry. The loss of peptide-pulsed target cells was indicative of cytotoxicity. The ratio of pulsed to unpulsed target cells in the indicated groups of mice was calculated and the percent killing was determined by using the formula: [1- (ratio in experimental mouse/ratio in naive mouse)] x 100%.

Murine skin transplantation: BALB/c mice served as skin donors and

C57BL/6 mice served as allograft recipients. Full-thickness skin grafts were harvested from the back and then fixed on the thoracic flank of recipient mice with simple separate stitches. The size of transplanted grafts was 1 ^ 1 cm 2 . Grafts were observed every day after the removal of the bandage at day 7 and considered rejected when > 90% of the graft tissue became necrotic. The MST of skin grafts was observed. Seven days after surgery, skin tissue from the transplantation site was collected for optical microscopy after hematoxylin and eosin staining.

Murine heterotopic heart transplantation: BALB/c mice served as heart donors and C57BL/6 mice serve as allograft recipients. Either abdominal or cervical heterotopic heart transplantation was performed according to previously published methods (Corry et al. (1973) Transplantation 16(4):343-350; Oberhube et al. (2014) J Vis Exp 92:e50753). Functionality of the transplanted heart was monitored daily by palpation. Clinical rejection was defined by cessation of palpable heartbeats and confirmed by autopsy. Loss of graft function within 48 hrs of transplantation was considered as a technical failure, and animals in which this occurred were omitted from the analysis.

Statistical analysis: Prism software version 5.0 (GraphPad Software) was used for statistical analyses, including unpaired Student's /-test, two-way analysis of variance (ANOVA) and log-rank analysis. A P value less than 0.05 was considered statistically significant.

Introduction

Advances in immunosuppressive regimens have played an essential role in driving forward the field of organ transplantation (Sayegh & Carpenter (2004) The New England Journal of Medicine 351(26):2761-2766). However, long-term use of immunosuppressants not only leads to susceptibility to infection, but also results in a broad range of co-morbidity. For example, calcineurin inhibitors are associated with hyperlipidemia, hyperglycemia, neuro- and nephrotoxicity, as well as an increased risk of malignancy (Crutchlow & Bloom (2007) Clinical Journal of the American Society of ' Nephrology: J4S 2(2):343-355; Hoorn et al. (2012) Journal of

Nephrology 25(3):269-275; Arnold et al. (20\3) Am J Transplant \3(9):2426-2432; Guba et al. (2004) Transplantation 77(12): 1777-1782; Roodnat e/ 1 a/. (2014)

Transplantation 98(l):47-53.2-6 ). In addition, such agents inhibit negative regulatory and tolerance-inducing responses (Wu et al. (2012) Transpl Immunol

27(l):30-38). That is, the calcineurin inhibitors are truly immunosuppressive in that they inhibit both activating and inhibitory signaling pathways (Powell & Zheng (2006) Curr Opin Investig Drugs 7(11): 1002-1007). As such, whereas the ultimate goal of anti -rejection strategies is to induce immune tolerance in the absence of long- term immunosuppression, current treatment regimens thwart this goal by inhibiting the induction of tolerance. Therefore new approaches to preventing graft rejection are required.

Recently, metabolic signaling pathways have been shown to play critical roles in dictating the outcomes of T cell responses (Yang & Chi (2012) Seminars in Immunology 24(6):421-428; Waickman & Powell (2012) Immunological Reviews

249(l):43-58; Pollizzi & Powell (2014) Nature Reviews. Immunology 14(7):435-446). The coordination of metabolism reprogramming and T cell function reflects the ability of how low-frequency antigen-specific naive T cells rapidly expand in response to a pathogen (Powell et al. (2013) Cold Spring Harbor Symposia on Quantitative Biology 78: 125-130). In the presence of oxygen, naive or resting T cells rely on mitochondrial oxidative phosphorylation (OXPHOS) to generate energy necessary for immune surveillance (Pearce et al. (2013) Science 342(6155): 1242454). In contrast, both CD4 + and CD8 + effector T cells employ aerobic glycolysis to meet their biosynthetic demands (Pearce et al. (2013) Science 342(6155): 1242454; Jones & Thompson (2007) Immunity 27(2): 173-178). This use of glycolysis in the presence of oxygen was first described by Otto Warburg in cancer cells (Warburg (1956) Science 124 (3215):269-270) and was subsequently found to be important in activated T cells (Warburg et al. (1958) [Metabolism of leukocytes]. Zeitschrift fur

Naturforschung. Teil B: Chemie, Biochemie, Biophysik, Biologie 13B (8):515-516). While glycolysis provides less adenosine triphosphate (ATP) than oxidative phosphorylation, it has been proposed that 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). Essential for this activation-induced glycolytic response is glucose uptake (Cham et al. (2008) European Journal of Immunology

38(9):2438-2450; Cham & Gajewski (2005) Journal of Immunology

174(8):4670-4677). Indeed, the increased expression of the glucose transporter GLUT1 on the cell surface is a critical aspect of TCR-induced activation (Jacobs et al. (2008) Journal of Immunology 180(7):4476-4486). Similarly, the uptake and metabolism of amino acids, especially glutamine, is essential for T cell activation (Carr et al. (2010) Journal of Immunology 185(2): 1037-1044). Glutamine deprivation blocks T cell proliferation and cytokine production (Carr et al. (2010) Journal of Immunology 185(2): 1037-1044). While the considerations depicted above reflect the metabolic needs of T cells during activation and explosive proliferation, what has also become apparent is that different T cell subsets require different metabolic programs. For example, Thl, Th2, and Thl7 effector T cells have been found to depend on glucose uptake and glycolysis. Alternatively, regulatory T cells (Tregs) are dependent upon lipid oxidation to generate energy (Michalek et al. (2011) Journal of

Immunology 186(6):3299-3303). Blocking glycolysis inhibits effector development but promotes Treg formation (Shi et al. (2011) The Journal of Experimental Medicine 208(7): 1367-1376). Furthermore, CD4 + effector T cells with GLUT1 deficiency are impaired in proliferation and function in vivo, whereas Tregs are enriched and functionally unaffected (Macintyre et al. (2014) CellMetab 20(l):61-72). Likewise, glutamine is critically required by naive CD4 + T cell to differentiate to Thl and Thl 7 effector T cells but not to Tregs (Nakaya et al. (2014) Immunity 40(5):692-705). Thus, strategies designed to inhibit metabolic reprogramming to prevent naive T cells from activating and differentiating into effector T cells, might provide a novel means to inhibit transplant rejection. In this study, it is demonstrated that combined inhibition of glycolysis and glutamine metabolism leads to a profound suppression in CD4 and CD8 effector T cell responses while preserving mechanisms of immune regulation. It is proposed that this anti-metabolic regimen represents a novel and promising strategy to promote allograft survival.

Results

Blocking glycolysis inhibits activation-induced T cell proliferation and cytokine production: To assess the effect of metabolic reprogramming agents on the metabolic changes of resting T cells in response to stimulation, the two intrinsic cellular bioenergetic parameters, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) with XF96 extracellular flux analyzer (Seahorse

Bioscience), were measured. OCR is an indicator of mitochondrial OXPHOS, and ECAR is predominantly a measure of lactic acid formed during glycolytic energy metabolism, and thus reflects overall glycolytic flux. Primary, naive 5C.C7 CD4 + T cells were incubated with pigeon cytochrome c (PCC) peptide for 48 hrs and then expanded and rested in IL-2 for an additional 5 days to generate previously activated CD4 + cells. After measuring the baseline bioenergetics of these resting CD4 + cells, cells were re-stimulated with anti-CD3/CD28 (FIG. 9A and FIG. 9B). Upon activation, the T cells exhibited a marked increase in ECAR and a modest increase in OCR in response to anti-CD3/CD28 (FIG. 9A and FIG. 9B). Upon reaching the peak of glycolytic flux, metabolic reprogramming agents were administered to the cells. 2- DG is a glucose analog that inhibits hexokinase, the first enzyme in the glycolytic pathway (Rowe et al. (2013) Nature Medicine 19(4):488-493). Adding 2-DG led to a sharp decrease of ECAR levels and an insignificant increase in OCR (FIG. 9A and FIG. 9B), reflecting that 2-DG inhibited glycolysis but minimally affected OXPHOS (Cheng et al. (2012) Cancer Research 72(10):2634-2644; Yamasaki et al. (2011) Nature Reviews. Urology 8(3): 165-171). Metformin is a commonly used oral hypoglycemic that activates AMPK, inhibits mitochondrial respiratory complex I and promotes fatty acid oxidation (Buzzai et al. (2007) Cancer Research 67(14):6745- 6752; El-Mir et al. (2000) The Journal of Biological Chemistry 275(l):223-228). Consistent with its ability to inhibit Complex I (El-Mir et al. (2000) The Journal of Biological Chemistry 275(l):223-228), the addition of metformin led to a marked suppression of OCR and an increase in ECAR (FIG. 9A and FIG. 9B). However, concomitant use of 2-DG and metformin together has been shown to potentiate the inhibition of glycolysis in tumor cells (Cheong et al. (2011) Molecular Cancer Therapeutics 10(12):2350-2362; Cheng et al. (2014) British Journal of ' Cancer 111(1):85-93.31). Indeed, when combined with metformin, 2-DG inhibits ECAR in T cells more effectively than when employed alone (FIG. 9A). In addition, the combination of 2-DG and metformin led to a lower OCR compared to that of the no treatment group but higher than that of metformin alone (FIG. 9B). Thus, similar to observations in tumor cells, combining 2-DG and metformin leads to a profound decrease in glycolysis in activated T cells.

Given the critical role of glycolysis in promoting T cell effector function, the effect of 2-DG and metformin on T cell proliferation and cytokine production was determined next. To this end, splenocytes from wild-type (WT) C57BL/6 mice were stimulated with anti-CD3 in the presence of media control, 2-DG alone, metformin alone or 2-DG + metformin for 72 hrs. Initial titration studies demonstrated that 2- DG metabolic reprogramming therapy or metformin metabolic reprogramming therapy inhibited T cell proliferation and cytokine production in a dose-dependent manner (data not shown). Importantly, consistent with their combined metabolic effects, the combination of 2-DG and metformin showed a more significant inhibition of cell proliferation and IFN-γ production of activated CD4 + and CD8 + T cells than either compound alone (FIG. 9C and FIG. 9D). Together, these data suggest that 2- DG in combination with metformin potently inhibit both metabolic reprogramming and proliferation/cytokine production in activated T cells.

Combined inhibition of glycolysis and glutaminolysis profoundly abrogates activation-induced T cell proliferation and cytokine production: Upon activation, glutamine oxidation increases as well as glycolysis to meet biosynthetic demand and allowing for continued tricarboxylic acid (TCA) flux (Maclver et al. (2013) Annual Review of Immunology 31 :259-283). Consequently, T cell activation, differentiation and function are critically dependent upon adequate glutamine availability (Carr et al. (2010) Journal of Immunology 185(2): 1037-1044; Nakaya ei a/. (2014) Immunity 40(5):692-705). To this end, upon activation, T cells coordinately increase the expression of glutamine transporters and glutaminase activity required for glutamine oxidation (Carr et al. (2010) Journal of Immunology 185(2) : 1037-1044). 6-Diazo-5- oxo-L-norleucine (DON) is a glutamine analog that inhibits multiple targets in the glutamine metabolic pathway, including glutaminase and glutamine transporters (Thomas et al. (2014) Biochemical and Bbiophysical Research Communications 443(l):32-36; Thomas et al. (2013) Biochemical and Biophysical Research Communications 438(2):243-248). To explore the role of glutaminase activity in T cell activation, naive CD4 + T cells were stimulated with anti-CD3/CD28 in medium with or without metabolic reprogramming agents for 24 hrs. The glutaminase activity of these CD4 + T cells was determined using a radiolabeled assay as described (Thomas et al. (2013) Biochemical and Biophysical Research Communications 438(2):243-248). It was observed that activated T cells exhibited increased glutaminase activity compared to naive T cells (FIG. 10A), indicating metabolic reprogramming toward glutaminolysis. Notably, glutaminase activity of activated T cells was significantly abrogated in the presence of DON (FIG. 10A), and this inhibition was not affected by the presence of the glycolytic inhibitors (FIG. 10A).

Furthermore, when cultured with DON, activated CD4 + and CD8 + T cells had decreased proliferation and IFN-γ production (FIG. 10B and FIG. IOC). Having confirmed previous studies showing that blocking the glutamine metabolic pathway was also capable of suppressing T cell responses (Carr et al. (2010) Journal of Immunology 185(2): 1037-1044), it was next investigated whether dual inhibition of glycolysis and glutaminolysis had an additional effect on inhibiting T cell proliferation and function. To this end, metabolic reprogramming agents 2-DG, metformin and DON were combined as a metabolic reprogramming therapy. Indeed, metabolic reprogramming therapy with at least three metabolic reprogramming agents had a more profound effect on suppressing T cell proliferation and IFN-γ production than only metabolic reprogramming therapy with at least two metabolic

reprogramming agents (e.g., 2-DG and metformin) or at least one metabolic reprogramming agent (e.g., DON)(FIG. 10B and FIG. IOC). Of note, the decrease in function was not because of a decrease of cell viability (FIG. 11). In addition, although glutamate can be further catabolized to fuel TCA cycle and ATP production (Pearce et al. (2013) Science 342(6155): 1242454), in extracellular flux analysis, adding DON (individually or combined with 2-DG and metformin) did not have an obvious impact on either OXPHOS or glycolytic flux of activated cells (FIG. 12A and FIG. 12B). Therefore, it was concluded that the inhibition of activation-induced T cell proliferation and function was most effective by simultaneously blocking glycolysis (with 2-DG and metformin) and glutaminolysis (with DON).

Inhibition of glycolysis and glutaminolysis suppresses the proliferation and function of antigen-specific CD4 + effector T cells but not Tregs in vivo: Given the observation that inhibition of glycolysis and glutaminolysis suppresses the activation of T cells in vitro, this paradigm was examined in an antigen-specific manner in vivo. To test the ability of metabolic reprogramming agents to mitigate CD4+ effector T cell responses, Thyl. l + CD4 + OVA-specific (OT-II) TCR transgenic T cells were adoptively transferred into Thyl.2 + WT mice that were concomitantly infected with vaccinia-OVA, a recombinant vaccinia virus carrying chicken ovalbumin (OVA) on day 0. The mice were then treated with PBS (vehicle control), 2-DG+metformin (metabolic reprogramming therapy with at least two metabolic reprogramming agents) or 2-DG+metformin+DON (metabolic reprogramming therapy with at least three metabolic reprogramming agents) for 3 days and the expansion of the OVA- specific Thy 1.1 + CD4 + T cells was interrogated on day 4. Mice treated with 2- DG+metformin demonstrated a marked decrease in the expansion of the antigen- specific Thyl. l + CD4 + T cells, which was even further reduced with the metabolic reprogramming therapy with at least three metabolic reprogramming agents (FIG. 13 A). In addition, the metabolic reprogramming therapy with at least three metabolic reprogramming agents in vivo strongly inhibited antigen-specific IFN-γ production of splenocytes upon re-challenge with peptide ex vivo (FIG. 13B). Metabolic reprogramming therapy with at least three metabolic reprogramming agents also led to a significantly higher frequency of Foxp3 + CD4 + T cells (FIG. 13C), indicating a preservation of this mechanism of immune regulation. Of note, the expansion and absolute number of Thy 1.1 + Foxp3 + T cells was decreased in the mice treated with metabolic reprogramming therapy with at least three metabolic reprogramming agents when compared to the untreated mice. This indicates that, as is the case for effector cells, the expansion of Foxp3 + T cells in response to antigen requires glycolysis and glutamine. However, the ratio of OVA-specific Thyl. l + regulatory T cells: Effector cells was increased in the treated mice (FIG. 13D). Together, these results showed that metabolic reprogramming therapy with at least three metabolic reprogramming agents had the most robust effect on inhibiting CD4 + T cell proliferation and cytokine production upon antigen recognition, while the differentiation into antigen-specific regulatory T cells was relatively preserved.

Inhibition of glycolysis and glutaminolysis suppresses the proliferation and function of antigen-specific CD8 + T cell: Next, the ability of the metabolic reprogramming agents to block antigen-specific CD8 + T cell responses was examined. To this end, Thyl. l + CD8 + OVA-specific (OT-I) TCR transgenic T cells were adoptively transferred into Thyl.2 + WT mice and the mice were infected with vaccinia-OVA and treated with metabolic reprogramming agents (the same regimen used in OT-II experiments). As was the case for CD4 + T cells, treatment of the mice with metabolic reprogramming therapy with at least three metabolic reprogramming agents led to a more profound inhibition of the expansion of the antigen-specific Thyl.l + CD8 + T cells than 2DG + metformin (FIG. 14A) or any compound alone (data not shown). Additionally, this combination of anti-metabolic metabolic reprogramming therapy strongly inhibited the generation of IFN-y-secreting CD8 + T cells (FIG. 14B). The decreased frequency and function of Thyl.l + CD8 + T cells was also reflected by the amount of IFN-γ secreted by splenocytes re-stimulated ex vivo with class I OVA peptide (FIG. 14C). Having demonstrated the ability of inhibitors of glycolysis and glutaminolysis to mitigate the expansion and function of exogenous effector T cells, the effect of the anti-metabolic regimen on endogenous effector CD8 + T cells was next examined with an in vivo cytotoxicity (CTL) assay. Mice were immunized with vaccinia-OVA and treated with PBS (vehicle control), metabolic reprogramming therapy with at least two metabolic reprogramming agents (e.g., 2DG + metformin) or metabolic reprogramming therapy with at least three metabolic reprogramming agents (e.g., 2DG + metforming + DON) for 7 days, then CFSE- labeled target cells were injected on day 8 and percent of specific killing was evaluated 10 hrs later. It was observed that most (96% ± 2%) of the OVA peptide- pulsed target cells were preserved in the mice receiving metabolic reprogramming therapy with at least three metabolic reprogramming agents, whereas 78 ± 5% and 45 ± 10% of the target cells were eliminated in mice receiving PBS and 2- DG+metformin treatment, respectively (FIG. 14D). These data suggest that the development of effective cytotoxic T lymphocytes is suppressed by the inhibition of glycolysis and glutamine metabolism.

Metabolic reprogramming agents promote allograft acceptance in mouse models of skin and heart transplantation: Having demonstrated the ability of combined anti-metabolic metabolic reprogramming therapy to inhibit anti-viral responses in vivo, it was next investigated whether anti-metabolic metabolic reprogramming therapy could prolong allograft survival using a skin transplantation model involving C57BL/6 mice receiving allogenic skin grafts from Balb/c mice. A significantly prolonged graft median survival time (MST) was found for mice that received 2-DG+metformin or DON alone, compared to those that received no treatment (13 vs. 11 days, P = 0.0048 and 22 vs. l ldays, P=0.0019, respectively) (FIG. 15 A). However, blocking at least the metabolic pathways of glycolysis and glutamine metabolism with metabolic reprogramming therapy with at least three metabolic reprogramming agents resulted in a marked increase of skin graft survival compared to no treatment (MST 40 vs. 11 days, P < 0.0001) (FIG. 15A). In addition, the appearance and histology of skin grafts from the group treated with metabolic reprogramming therapy with at least three metabolic reprogramming agents exhibited no evidence of tissue destruction, more intact tissue alignment, and less infiltration of inflammatory cells (FIG. 15B, FIG. 15C, FIG. 15D, and FIG. 15E). For comparison, the MST of this fully allogeneic skin allograft under the treatment of FK506 or cyclosporine has previously been reported to be less than 20 days (Gorski & Wasik (1990) Immunology 71(1): 148-150).

Finally, this metabolic reprogramming therapy with at least three metabolic reprogramming agents was employed in a Balb/c to C57BL/6 heart transplantation, which is a vascularized solid organ transplantation model. Strikingly, when recipient mice were treated with metabolic reprogramming therapy with at least three metabolic reprogramming agents, the grafted hearts continued beating for more than 100 days post-transplantation without rejection (FIG. 15F). Likewise, the cardiac grafts from the treatment group exhibited much healthier microscopic morphology with no evidence of necrosis, fibrosis, or lymphocyte infiltration (FIG. 15G, FIG. 15H, FIG. 151 and FIG. 15 J). Together, these data support the notion that the inhibition of glycolysis and glutamine metabolic pathways represents a potent means to prevent acute rejection and promote long-term graft acceptance.

Discussion

In this study a novel approach was defined to prevent transplantation graft rejection by inhibiting metabolic pathways necessary for effector T cell function. This approach is based upon the emerging understanding that metabolic

reprogramming in T cells is not simply the result of T cell activation, but rather plays a critical role in regulating T cell differentiation and function (Jones & Thompson (2007) Immunity 27(2): 173-178; Maclver et al. (2013) Annual Review of Immunology 31 :259-283). The therapeutic index of this approach relies on the extraordinary metabolic requirements of effector T cell response that is distinct from the more flexible and adaptive ordinary metabolic network. As such, even though the inhibitors employed are relatively non-specific, relative cellular selectivity is achieved based upon the extraordinary metabolic demands of the effector T cells. Indeed, while effector T cells employ aerobic glycolysis for energy, regulatory T cells are programmed such that they rely upon oxidative phosphorylation and fatty acid oxidation (Michalek et al. (2011) Journal of Immunology 186(6):3299-330322). Nonetheless, in this study, inhibiting glycolysis by the combination of 2-DG and metformin was not sufficient to prevent acute rejection or induce long-term tolerance in a fully MHC-mismatched transplantation model. Inhibition of glutamine metabolism also blocks effector T cell differentiation, proliferation and function while leaving Treg differentiation relatively intact (Carr et al. (2010) Journal of

Immunology 185(2): 1037-1044; Nakaya ei a/. (2014) Immunity 40(5):692-705). Thus, DON, a glutaminolytic inhibitor, was integrated into this protocol. As such, inhibiting both glycolysis and glutaminolysis, (a regimen that can simultaneously suppress effector T cell function while preserving Treg differentiation) proved to be the most effective in promoting graft survival (Shi et al. (2011) The Journal of Experimental Medicine 208(7): 1367-1376; Michalek et al. (2011) Journal of Immunology 186(6):3299-3303).

The cellular selectivity based on demand was not only observed for cells of the immune system but also contributed to the relative lack of side effects observed. Indeed, by tracking body weight and fur ruffling as markers of overall health of transplant recipient mice, in some cases for more than 100 days, any systemic sickness or morbidity was not observed associated with the treatment regimen.

Further, the compounds employed in the study have all been employed in patients with tolerable adverse effects (Franciosi et al. (2013) PloS one 8(8):e71583; Sullivan et al. (1988) Cancer Chemometabolic reprogramming therapy and Pharmacology 21(l):78-84; Raez et al. (2013) Cancer Chemometabolic reprogramming therapy and Pharmacology 71(2):523-530). Thus, it is believed that the combination treatment strategy is readily translated into the clinic. Notably, in previous trials using these 2- DG or DON or metformin for the treatment of cancer, the efficacy was not as striking as what has been observed in using these agents for transplant rejection (Sullivan et al. (1988) Cancer Chemometabolic reprogramming therapy and Pharmacology 21(l):78-84; Raez et al. (2013) Cancer Chemometabolic reprogramming therapy and Pharmacology 71(2):523-530; Tsilidis et al. (2014) Diabetes Care 37(9):2522-2532). It is believed that this is because while T effector cells and cancer cells both demonstrate Warburg metabolism, mutations in tumors enable them to more effectively become resistant to these agents. Indeed, it is believed that the enhanced sensitivity of T cells to the combination regimen contributes to the ability to devise a regimen with doses that have relatively minor side effects.

Steroids and calcineurin inhibitors remain the backbone of most current posttransplantation regimens. Steroids have multiple adverse effects, such as increased risk of infection, hyperglycemia, accelerated atherosclerosis, and gastrointestinal bleeding, while calcineurin inhibitors are associated with neurotoxicity and nephrotoxicity, as well as risk of infection and an increased risk of cancer (Crutchlow & Bloom (2007) Clinical Journal of the American Society of Nephrology: CJASN 2(2):343-355; Hoorn et al. (2012) Journal of Nephrology 25(3):269-275; Arnold et al. (2013) Am J Transplant 13(9):2426-2432; Guba et al. (2004) Transplantation

77(12): 1777-1782; Roodnat et a/. (2014) Transplantation 98(l):47-53.2-6). The use of the presently disclosed agents is devoid of the side effects associated with current immunosuppressive regimens. At least one advantage of metabolic reprogramming therapy is that it will promote a favorable systemic metabolic profile. Indeed, the metabolic reprogramming agents employed counteract some of the metabolic abnormalities associated with current immunosuppression, such as increased blood glucose and triglycerides. Of note however, one of the major obstacles of current transplantation immunosuppression is the reactivation of cytomegalovirus.

Interestingly, the agents employed in the anti-metabolic approach have been shown to inhibit viral replication (Chambers et al. (2010) J Virol 84(4): 1867-1873). In addition, while calcineurin inhibitors are associated with an increased risk of developing neoplasm (Guba et al. (2004) Transplantation 77(12): 1777-1782, targeting both glycolysis and glutamine pathway inhibits the growth of tumor cells (Cheong et al. (2011) Molecular Cancer Therapeutics 10(12):2350-2362; Cheng et al. (2014) British Journal of Cancer 111(1):85-93.31; Willems et al. (2013) Blood 122(20):3521-3532; Lim et al. (2014) Cancer Research 74(13):3535-3545).

Furthermore, while calcineurin inhibitors have efficacy in the prevention of acute rejection, such agents can block the induction of immunologic tolerance in part by preventing T cell anergy and inhibiting the generation of regulatory T cells. The metabolic reprogramming agents employed in this study promote both anergy and T regulatory cell generation (FIG. 13C; Zheng et al. (2009) Journal of Immunology 183(10):6095-6101). However, metabolic reprogramming metabolic reprogramming therapy alone was not sufficient to completely prevent skin allograft rejection.

Further, even though 100% heart graft survival was observed (FIG. 15D) while the mice were being treated, preliminary studies indicate that stopping metabolic reprogramming therapy resulted in the eventual rejection of the hearts approximately 80 days later (data not shown). Integrating tolerance inducing metabolic

reprogramming therapy, such as costimulatory blockade with, or without, rapamycin, may be a means to induce long-term tolerance in the absence of long-term

immunosuppression (Oderup et al. (2006) Transplantation 82(11): 1493-1500; Pilon et al. (2014) American Journal of Transplantation: Official Journal of the American Society of Transplantation and the American Society of Transplant Surgeons

14(12):2874-2882; Bestard ei a/. (2011) Transplant International: Official Journal of the European Society for Organ Transplantation 24(5): 451-460).

Studies have revealed a critical role for the mammalian Target Of Rapamcyin (mTOR) in regulating T cell differentiation and function (Delgoffe et al. (2011) Nature Immunology 12(4): 295 -303; Li et al. (2011) Immunity 34(4):541-553). By genetically dissecting mTOR signaling in mice, TCR engagement in the absence of mTOR was demonstrated to lead to the generation of Tregs (Delgoffe et al. (2011) Nature Immunology 12(4):295-303; Delgoffe et al. (2009) Immunity 30(6):832-844). However, in spite of these revealing genetic studies, the efficacy of rapamycin and other rapalogues in preventing transplantation rejection has been somewhat pedestrian (Abdelmalek et al. (2012) American Journal of Transplantation: Official Journal of the American Society of Transplantation and the American Society of Transplant Surgeons 12(3): 694-705). In part, it is believed that this is due to a lack of strategic integration of mTOR inhibitors into immunosuppressive protocols (Lo et al. (2014) Current Opinion in Organ Transplantation 19(4):363-371). For example, combining rapamycin with calcineurin inhibitors negate the potentially tolerance inducing effects of rapamycin (Satake et al. (2014) PloS one 9(3):e9288854). Interestingly, what has emerged from recent studies is that mechanistically, mTOR regulates T cell differentiation and function in part by regulating metabolic programs. mTOR plays an important role in regulating glycolysis and glutamine metabolism which are critical in order to support effector T cell generation and function (Wang et al. (2011) Immunity 35(6):871-882; Sengupta et al. (2010) Molecular Cell 40(2): 310-322; Csibi et al. (2013) Cell 153(4):840-854). Thus, inhibition of mTOR activity abrogates effector T cell generation (both CD4 + and CD8 + ) and also hinders the up-regulation of glycolysis and glutaminolysis upon T cell activation. The presently disclosed metabolic reprogramming metabolic reprogramming therapy approach directly targets these pathways.

In conclusion, the presently disclosed subject matter provides a novel approach to prevention of graft rejection by inhibiting metabolic pathways necessary for effector T cell function. The differential metabolic requirements of effector and regulatory T cells reveal a new therapeutic window to simultaneously inhibit rejection and promote tolerance. Future studies combining metabolic reprogramming metabolic reprogramming therapy with tolerance-inducing regimens, such as co- stimulatory blockade and Treg metabolic reprogramming therapy, have the potential to promote long-term graft acceptance in the absence of long-term

immunosuppression.

EXAMPLE 3

Mice were sensitized to House Dust Mite antigen (HDM) in the absence of drug. Upon intratracheal rechallenge the mice were treated with vehicle or 25. In this model during the acute lung rechallenge 25 inhibited pathology, the

recruitment/generation of Th2 cells and reduced the levels of HDM specific IgE (FIG. 16). This data shows the effectiveness of using a DON prodrug to treat asthma.

EXAMPLE 4

FIG. 17A, FIG. 17B, and FIG. 17C show targeting glutamine in a mouse model of acute respiratory distress syndrome (ARDS). Mice were challenged with Ips (FIG. 17A) to induce ARDS and treated with DON on day 2 & 4. Treatment promoted a more rapid recovery (weight gain) (FIG. 17B) and less lung damage (FIG. 17C). This data shows the effectiveness of using DON to treat ARDS.

EXAMPLE 5

Mice were treated with intratracheal bleomycin to induce fibrosis. DON treated mice demonstrated markedly decreased lung damage as well as a decrease in the recruitment/generation of lung fibrocytes (FIG. 18). This data shows the effectiveness o fusing DON to treat pulmonary fibrosis.

EXAMPLE 6

FIG. 19A and FIG. 19B show the targeting of glutamine metabolism to cure Cerebral malaria. Mice were treated on day 5 post infection and on day 6 (12 hours before subsequent death) with DON every other day. The striking ability of DON to prevent death at such a late stage of disease highlights the robust ability of our approach in CNS inflammation.

EXAMPLE 7

FIG. 20 shows the targeting of glutamine metabolism in neuromyelitis optica. Auto-reactive T cells were adoptively transferred into mice treated with PBS or 25 and mean paralysis scores were measured. This data shows the effectiveness of using a DON prodrug to treat neuromyelitis optica.

EXAMPLE 8

FIG. 21 A and FIG. 2 IB show the targeting of glutamine metabolism in multiple sclerosis. Animals were immunized for EAE (C57BL/6+MOG35-55). Mice were treated with vehicle or 25 either q.d. from the day of immunization (day 0, left), or b.d. from the time of disease onset (EAE disease score >1, right). This data shows the effectiveness of using a DON prodrug to prevent (FIG. 21 A) and treat (FIG. 2 IB) multiple sclerosis.

EXAMPLE 9

FIG. 22 shows screening prodrugs in vivo: Thy 1.1+ OT-1 cells were adoptively transferred into mice infected with vaccinia ova. The mice were treated with prodrugs or vehicle (V) and the ova-specific response was monitored. In this case prodrugs PI, PI -2 and P3 effectively inhibited the T cell response. REFERENCES

All publications, patent applications, patents, and other references mentioned in the specification are indicative of the level of those skilled in the art to which the presently disclosed subject matter pertains. All publications, patent applications, patents, and other references are herein incorporated by reference to the same extent as if each individual publication, patent application, patent, and other reference was specifically and individually indicated to be incorporated by reference. It will be understood that, although a number of patent applications, patents, and other references are referred to herein, such reference does not constitute an admission that any of these documents forms part of the common general knowledge in the art. In case of a conflict between the specification and any of the incorporated references, the specification (including any amendments thereof, which may be based on an incorporated reference), shall control. Standard art-accepted meanings of terms are used herein unless indicated otherwise. Standard abbreviations for various terms are used herein.

Although the foregoing subject matter has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be understood by those skilled in the art that certain changes and modifications can be practiced within the scope of the appended claims.