US20140030295A1 - Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response - Google Patents

Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response Download PDF

Info

Publication number
US20140030295A1
US20140030295A1 US13/717,041 US201213717041A US2014030295A1 US 20140030295 A1 US20140030295 A1 US 20140030295A1 US 201213717041 A US201213717041 A US 201213717041A US 2014030295 A1 US2014030295 A1 US 2014030295A1
Authority
US
United States
Prior art keywords
compound
sirt1
individual
cell
inhibitor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/717,041
Inventor
Hyungwook Lim
Melanie Ott
Eric M. VERDIN
Hye-Sook Kwon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
J David Gladstone Institutes
Original Assignee
J David Gladstone Institutes
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by J David Gladstone Institutes filed Critical J David Gladstone Institutes
Priority to US13/717,041 priority Critical patent/US20140030295A1/en
Assigned to THE J. DAVID GLADSTONE INSTITUTES reassignment THE J. DAVID GLADSTONE INSTITUTES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KWON, HYE-SOOK, LIM, HYUNGWOOK, OTT, MELANIE, VERDIN, ERIC M.
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: J. DAVID GLADSTONE INSTITUTES
Publication of US20140030295A1 publication Critical patent/US20140030295A1/en
Assigned to THE J. DAVID GLADSTONE INSTITUTES, A TESTAMENTARY TRUST ESTABLISHED UNDER THE WILL OF J. DAVID GLADSTONE reassignment THE J. DAVID GLADSTONE INSTITUTES, A TESTAMENTARY TRUST ESTABLISHED UNDER THE WILL OF J. DAVID GLADSTONE CORRECTIVE ASSIGNMENT TO CORRECT THE NAME TO THE J. DAVID GLADSTONE INSTITUTES, A TESTAMENTARY TRUST ESTABLISHED UNDER THE WILL OF J. DAVID GLADSTONE PREVIOUSLY RECORDED ON REEL 029730 FRAME 0804. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: KWON, HYE-SOOK, LIM, HYUNGWOOK, OTT, MELANIE, VERDIN, ERIC M.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • Immune activation is a hallmark of human immunodeficiency virus-1 (HIV-1) infection and a significant factor that promotes continuous viral replication and CD4+ T-cell depletion.
  • HIV-1 human immunodeficiency virus-1
  • levels of circulating activation markers correlate with accelerated disease progression and shortened survival.
  • HIV infection is critically dependent on the activated state of CD4+ T cells since the virus cannot replicate efficiently in resting T cells.
  • Quiescent T cells in blood are refractory to infection because of blocks at the level of reverse transcription and proviral integration.
  • T-cell activation enhances viral transcription through the activation of various transcription factors, notably nuclear factor ⁇ B (NF- ⁇ B).
  • SIRT1 is a mammalian homologue of the yeast transcriptional repressor silent information regulator 2 (Sir2), an important factor governing longevity in yeast. Like Sir2, SIRT1 requires nicotinamide adenine dinucleotide (NAD + ) as a cofactor, which links its activity to the metabolic state of the cell. In addition to its enzymatic activity on histone substrates in vitro, recent experimental evidence suggests that SIRT1 predominantly targets nonhistone proteins for deacetylation. It has been reported that Tat is a substrate for the deacetylase activity of SIRT1.
  • Acetylation of lysine 50 (K50) in Tat is mediated by the histone acetyltransferase activities of p300 and human GCNS and generates binding sites for the bromodomains of PCAF and Brg1.
  • SIRT1 binds and deacetylates Tat at K50, a process necessary to recycle nonacetylated Tat protein for binding to TAR RNA and the cellular positive transcription elongation factor b (P-TEFb).
  • the present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an inhibitor of SIRT1.
  • the present disclosure provides a method of reducing an immune response, e.g., to treat chronic immune hyperactivity, the method generally involving administering to an individual in need thereof an activator of SIRT1.
  • the present disclosure provides a method of modulating activation and differentiation of CD4 + T cells.
  • FIGS. 1A-F depict data showing the effect of Tat and nicotinamide on T cell hyperactivation.
  • FIGS. 2A-D depict the Tat-mediated superinduction of NF- ⁇ B-responsive genes.
  • FIGS. 3A-C depicts the effect of Tat on the inhibitory effect of SIRT1 on NK- ⁇ -B-responsive promoters.
  • FIGS. 4A-E depict binding of Tat to the acetyl lysine-binding site in SIRT1.
  • FIGS. 5A-D depict the effect of Tat on SIRT1 deacetylase activity.
  • FIGS. 6A-C depict the effect of hyperacetylation of p65 by Tat on T-cell hyperactivation.
  • FIGS. 7A and 7B depict a model for Tat effects on SIRT1 deacetylase activity, T-cell activation, and HIV transcription.
  • FIGS. 8A and 8B depict the effect of SIRT1 activators on Tat-mediated T cell hyperactivation.
  • FIGS. 9A and 9B depict the effect of SIRT1 on induced Treg (iTreg) differentiation.
  • FIGS. 10A-D depict interaction of SIRT1 with FoxP3, and the effect of inhibition of SIRT1 deacetylase activity on acetylation of FoxP3.
  • FIGS. 11A-D depict destabilization of FoxP3 protein via deacetylation by SIRT1.
  • FIGS. 12A and 12B depict the effect of inhibition of SIRT1 activity on differentiation of iTreg cells and FoxP3 expression in nTreg cells.
  • FIG. 13 depicts an alignment of amino acid sequences of human FoxP3 (GenBank Accession Nos. NP — 054728 and NM — 014009), mouse FoxP3 (GenBank Accession Nos. NM — 054039 and NP — 473380), and cow FoxP3 (GenBank Accession Nos. NM — 001045933 and NP — 001039398).
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, e.g., in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, e.g., causing regression of the disease, e.g., to completely or partially remove symptoms of the disease.
  • an effective amount or “therapeutically effective amount” means a dosage sufficient to provide for treatment for the disease state being treated or to otherwise provide the desired effect (e.g., induction of an effective immune response, reduction of chronic immune hyperactivity, etc.).
  • the precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., weight, age, etc.), the disease, and the treatment being effected.
  • an individual refers to a mammal, including, but not limited to, murines, felines, simians, humans, etc.
  • an individual is a human.
  • an individual is a rodent (e.g., a mouse, a rat, etc.) or a lagomorph.
  • a “pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” and “pharmaceutically acceptable adjuvant” means an excipient, diluent, carrier, or adjuvant that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, or adjuvant that is acceptable for veterinary use as well as human pharmaceutical use.
  • “A pharmaceutically acceptable excipient, diluent, carrier and adjuvant” as used in the specification and claims includes one and more than one such excipient, diluent, carrier, and adjuvant.
  • a “pharmaceutical composition” is meant to encompass a composition suitable for administration to a subject, such as a mammal, e.g., a human.
  • a “pharmaceutical composition” is sterile, and is free of contaminants that are capable of eliciting an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade).
  • Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, intratracheal and the like.
  • the composition is suitable for administration by a transdermal route, using a penetration enhancer other than dimethylsulfoxide (DMSO).
  • DMSO dimethylsulfoxide
  • the pharmaceutical compositions are suitable for administration by a route other than transdermal administration.
  • a pharmaceutical composition will in some embodiments include a compound and a pharmaceutically acceptable excipient.
  • a pharmaceutically acceptable excipient is other than DMSO.
  • “pharmaceutically acceptable derivatives” of a compound of the invention include salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives may be readily prepared by those of skill in this art using known methods for such derivatization. The compounds produced may be administered to animals or humans without substantial toxic effects and are either pharmaceutically active or are prodrugs.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid
  • SIRT1 modulator includes a plurality of such modulators and reference to “the SIRT1 inhibitor” includes reference to one or more SIRT1 inhibitors and equivalents thereof known to those skilled in the art, and so forth.
  • the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation.
  • the present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an inhibitor of SIRT1.
  • the present disclosure provides a method of reducing an immune response (e.g., to treat chronic immune hyperactivity, to treat an autoimmune disorder), the method generally involving administering to an individual in need thereof an activator of SIRT1.
  • the present disclosure provides a method of modulating activation and differentiation of CD4 + T cells, the method generally involving contacting the CD4 + T cell with a SIRT1 inhibitor.
  • SIRT1 refers to a polypeptide exhibiting nicotinamide adenosine dinucleotide (NAD)-dependent deacetylase activity, and having an amino acid sequence that is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, identical to the amino acid sequence of a known SIRT1 polypeptide.
  • a SIRT1 protein includes yeast Sir2 (GenBank Accession No. P53685); C. elegans Sir-2.1 (GenBank Accession No. NP — 501912); human SIRT1 (see, e.g., GenBank Accession No.
  • NM — 012238 or NP — 036370 (or AF083106); Frye ((1999) Biochem. Biophys. Res. Comm. 260:273); and GenBank Accession Nos. Q96EB6, AAH12499, NP — 036370, and AAD40849); mouse SIR1 (see, e.g., GenBank Accession Nos. Q923E4 and NP — 062786); and equivalents and fragments thereof.
  • a SIRT1 polypeptide comprises an amino acid sequence having at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, amino acid sequence identity with a contiguous stretch of from about 500 amino acids (aa) to about 550 aa, from about 550 aa to about 600 aa, from about 600 aa to about 650 aa, from about 650 aa to about 700 aa, or from about 700 aa to about 747 aa, of the amino acid sequence set forth in SEQ ID NO:1.
  • a suitable SIRT1 modulator modulates an enzymatic activity of a SIRT1 polypeptide.
  • a suitable SIRT1 modulator modulates Tat deacetylase activity of SIRT1.
  • SIRT1 Tat deacetylase activity can be determined using known methods.
  • a Tat polypeptide e.g, a synthetic Tat polypeptide
  • a reaction sample can include an acetylated Tat polypeptide, NAD + , a SIRT1 polypeptide, and a SIRT1 modulator.
  • Acetylated Tat polypeptides include Tat polypeptides having one or more acetylated lysine residues, e.g., acetylated K28, acetylated K41, and acetylated K50.
  • a suitable acetylated Tat polypeptide includes the amino acid sequence Ser-Tyr-Gly-Arg-AcLys-Lys-Lys-Arg-Arg-Gln-Arg (SEQ ID NO:02), where AcLys is acetylated lysine.
  • An acetylated Tat protein can be generated as described in, e.g., Doerr et al. (2002) EMBO J. 21:2715-2723; or Peloponese (1999) J. Biol. Chem. 274:11473-11478.
  • Suitable acetylated Tat polypeptides are described in, e.g., U.S. Pat. No. 7,485,416 and U.S. Patent Publication No. 2005/0287597.
  • the effect of the SIRT1 modulator on SIRT1 Tat deacetylase activity can be determined by measuring the amount of deacetylated Tat polypeptide produced by action of the SIRT1 polypeptide in the presence of the SIRT1 modulator, compared to the amount of deacetylated Tat polypeptide produced by action of the SIRT1 polypeptide in a control reaction sample that does not include the SIRT1 modulator.
  • the amount of remaining acetylated Tat can be measured.
  • Methods of determining the level of acetylated Tat in a sample include immunological assays using antibody that is specific for acetylated form of Tat, and that therefore distinguishes between acetylated Tat and deacetylated Tat.
  • immunological assays can be used, including, e.g., enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), protein blot (“Western” blot) assays, and the like.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • protein blot (“Western” blot) assays, and the like.
  • mass spectroscopy is used. SIRT1 activity can also be determined by measuring the level of NAD in the test sample.
  • SIRT1 on acetylated Tat can be coupled to a second enzymatic reaction that reduces NAD to NADH, and measuring fluorescence of NADH at, e.g., 340 nm. See U.S. Pat. No. 7,485,416 and U.S. Patent Publication No. 2005/0287597 for descriptions of methods of determining SIRT1 Tat deacetylase activity.
  • a suitable SIRT1 modulator (a SIRT1 activator or a SIRT1 inhibitor) is a selective SIRT1 modulator.
  • a suitable SIRT1 activator is a selective SIRT1 activator; and in some embodiments, a suitable SIRT inhibitor is a selective SIRT1 inhibitor.
  • a suitable SIRT1 activator increases the enzymatic activity of a SIRT1 polypeptide, but does not substantially increase the enzymatic activity of any other sirtuin.
  • a suitable SIRT1 activator increases the enzymatic activity of a SIRT1 polypeptide, but does not substantially increase the enzymatic activity of SIRT2, SIRT4, or SIRT5.
  • a suitable SIRT1 inhibitor reduces the enzymatic activity of a SIRT1 polypeptide but does not substantially reduce the enzymatic activity of any other sirtuin.
  • a suitable SIRT1 inhibitor reduces the enzymatic activity of a SIRT1 polypeptide, but does not substantially reduce the enzymatic activity of SIRT2, SIRT4, or SIRT5.
  • a suitable SIRT1 modulator modulates the activity of SIRT1, and can also modulate the activity of one or more additional sirtuins.
  • a suitable SIRT1 activator increases the enzymatic activity of a SIRT1 polypeptide, and can also increase the enzymatic activity of SIRT5.
  • a suitable SIRT1 inhibitor reduces the enzymatic activity of a SIRT1 polypeptide, and can also reduce the enzymatic activity of SIRT5.
  • a suitable SIRT1 activator can increase the enzymatic activity of SIRT1 by at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 20-fold, or more than 20-fold.
  • a suitable SIRT1 activator can increase the enzymatic activity of SIRT1 by from about 25% to about 50%, from about 50% to about 75%, from about 75% to about 2-fold, from about 2-fold to about 5-fold, from about 5-fold to about 10-fold, from about 10-fold to about 20-fold, or more than 20-fold.
  • a suitable SIRT1 inhibitor can reduce the enzymatic activity of SIRT1 by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or more than 75%.
  • a suitable SIRT1 inhibitor can reduce the enzymatic activity of SIRT1 by from about 5% to about 10%, from about 10% to about 15%, from about 15% to about 20%, from about 20% to about 25%, from about 25% to about 30%, from about 35% to about 40%, from about 40% to about 50%, from about 50% to about 60%, from about 60% to about 75%, or more than 75%.
  • a suitable SIRT1 activator can increase SIRT1 enzymatic activity at an EC 50 (half maximal effective concentration) of from about 1 nM to about 1 mM, e.g., from about 1 nM to about 10 nM, from about 10 nM to about 15 nM, from about 15 nM to about 25 nM, from about 25 nM to about 50 nM, from about 50 nM to about 75 nM, from about 75 nM to about 100 nM, from about 100 nM to about 150 nM, from about 150 nM to about 200 nM, from about 200 nM to about 250 nM, from about 250 nM to about 300 nM, from about 300 nM to about 350 nM, from about 350 nM to about 400 nM, from about 400 nM to about 450 nM, from about 450 nM to about 500 nM, from about 500 nM to about 750 nM, from about 750 nM to
  • a suitable SIRT1 activator can increase SIRT1 enzymatic activity at an EC 1.5 (concentration of compound required to increase enzyme activity by 50%) of from about 1 nM to about 1 mM, e.g., from about 1 nM to about 10 nM, from about 10 nM to about 15 nM, from about 15 nM to about 25 nM, from about 25 nM to about 50 nM, from about 50 nM to about 75 nM, from about 75 nM to about 100 nM, from about 100 nM to about 150 nM, from about 150 nM to about 200 nM, from about 200 nM to about 250 nM, from about 250 nM to about 300 nM, from about 300 nM to about 350 nM, from about 350 nM to about 400 nM, from about 400 nM to about 450 nM, from about 450 nM to about 500 nM, from about 500 nM to about 750 nM, from
  • a suitable SIRT1 activator can have an EC 1.5 of from about 0.01 ⁇ M to about 100 ⁇ M, e.g., from about 0.01 ⁇ M to about 0.1 ⁇ M, from about 0.1 ⁇ M to about 0.5 ⁇ M, from about 0.5 ⁇ M to about 1.0 ⁇ M, from about 1.0 ⁇ M to about 10 ⁇ M, from about 10 ⁇ M to about 25 ⁇ M, from about 25 ⁇ M to about 50 ⁇ M, or from about 50 ⁇ M to about 100 ⁇ M.
  • a suitable SIRT1 inhibitor can have an IC 50 of less than 50 ⁇ M, e.g., a suitable SIRT1 inhibitor can have an IC 50 of from about 50 ⁇ M to about 5 nm, or less than 5 nM.
  • a suitable SIRT1 inhibitor has an IC 50 of from about 50 ⁇ M to about 25 ⁇ M, from about 25 ⁇ M to about 10 ⁇ M, from about 10 ⁇ M to about 5 ⁇ M, from about 5 ⁇ M to about 1 ⁇ M, from about 1 ⁇ M to about 500 nM, from about 500 nM to about 400 nM, from about 400 nM to about 300 nM, from about 300 nM to about 250 nM, from about 250 nM to about 200 nM, from about 200 nM to about 150 nM, from about 150 nM to about 100 nM, from about 100 nM to about 50 nM, from about 50 nM to about 30 nM, from about 30 nM, from about
  • SIRT1 modulators are known in the art, and any modulator of SIRT1 can be used.
  • Ota et al. ((2006) Oncogene 25:176) discusses sirtinol; resveratrol is discussed in, e.g., de la Lastra (2005) Mol. Nutr. Food Res. 49:405; Napper et al. ((2005) J. Med. Chem. 48:8045) discusses indole compounds that are SIRT1 inhibitors;
  • Solomon et al. ((2006) Mol. Cell. Biol. 26:28) discusses EX-527, a SIRT1 inhibitor;
  • U.S. Patent Publication Nos. 2007/0043050, 2007/0037865, and 2007/0037809 discuss SIRT1 modulators; and
  • U.S. Patent Publication No. 2008/0021063 discusses SIRT1 modulators.
  • a given agent e.g., a SIRT1 modulator, such as a SIRT1 inhibitor or a SIRT1 activator
  • an immune response e.g., increasing an immune response; e.g., reducing an immune response, such as reducing chronic immune hyperactivity
  • Immune response parameters include, but are not limited to, CD4 count; serum cytokine levels, e.g., serum levels of IL-2; serum antibody levels; levels of an autoantibody; and the like.
  • the present disclosure provides methods of reducing an immune response, the methods generally involving administering to an individual in need thereof an effective amount of a SIRT1 activator.
  • an effective amount of a SIRT1 activator is an amount that, when administered to an individual in one or more doses, is effective to reduce the level of a cytokine associated with an immune response.
  • Cytokines associated with an immune response include, e.g., interleukin-2 (IL-2), tumor necrosis factor-alpha (TNF- ⁇ ), interleukin-4 (IL-4), interferon gamma (IFN- ⁇ ), and interleukin-12 (IL-12).
  • an effective amount of a SIRT1 activator is an amount that, when administered to an individual in one or more doses, is effective to reduce the level in the individual of a cytokine associated with an immune response by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%, compared to the level of the cytokine in the absence of treatment with the SIRT1 activator.
  • an effective amount of a SIRT1 activator is an amount that, when administered to an individual in one or more doses, is effective to reduce the level of IL-2 (e.g., circulating IL-2, e.g., a serum level of IL-2) in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%, compared to the level of IL-2 in the individual in the absence of treatment with the SIRT1 activator.
  • IL-2 e.g., circulating IL-2, e.g., a serum level of IL-2
  • the instant disclosure provides methods for treating chronic immune hyperactivation, the methods generally involving administering to an individual in need thereof an effective amount of a SIRT1 activator.
  • the instant disclosure provides a method for treating chronic immune hyperactivation, the method comprising administering to an individual in need thereof an effective amount of a selective SIRT1 activator.
  • the instant disclosure provides a method for treating chronic immune hyperactivation that results from infection with an immunodeficiency virus, e.g., a human immunodeficiency virus (e.g., HIV-1), the method comprising administering to an individual in need thereof an effective amount of a SIRT1 activator, where in some embodiments, the SIRT1 inhibitor is a selective SIRT1 activator.
  • an immunodeficiency virus e.g., a human immunodeficiency virus (e.g., HIV-1)
  • the method comprising administering to an individual in need thereof an effective amount of a SIRT1 activator, where in some embodiments, the SIRT1 inhibitor is a selective SIRT1 activator.
  • SIRT1 activators that are suitable for use in a subject method include, but are not limited to, resveratrol ((E)-5-(p-Hydroxystyryl)resorcinol (E)-5-(4-hydroxystyryl)benzene-1,3-diol); or 3,5,4′-trihydroxy-trans-stilbene); butein (3,4,2′,4′-tetrahydroxychalcone); piceatannol (3,5,3′,4′-tetrahydroxy-trans-stilbene); isoliquiritigenin (4,2′,4′-trihydroxychalcone); fisetin (3,7,3′,4′-tetrahydroxyflavone); quercetin (3,5,7,3′,4′-pentahydroxyflavone); a SIRT1 activator as described in U.S.
  • a suitable SIRT1 activator is a compound of any one of Formulas I-XXVIII as described in U.S. Pat. No. 7,345,178, where substituents are as described in U.S. Pat. No. 7,345,178, or a pharmaceutically acceptable salt of a compound of any one of Formulas I-XXVIII as described in U.S. Pat. No. 7,345,178, provided that the compound activates SIRT1 activity.
  • a suitable SIRT1 activator includes a compound shown in Table 4 of U.S. Pat. No. 7,345,178.
  • SIRT1 activators are shown in Table 1, below.
  • SIRT1 activators include, e.g.,
  • SIRT1 activators include, e.g.,
  • SRT1720, SRT1460, and SRT2183 are selective SIRT1 activators. See, e.g., Milne et al. (2007) Nature 450:712.
  • SIRT1 activators that are quinoxaline compounds.
  • Suitable quinoxaline SIRT1 activators include, e.g., 3-benzenesulfonyl-1-(4-fluoro-phenyl)-1H-pyrrolo[2,3-b]quinoxalin-2-ylamine; 2-amino-1-(2-ethyl-phenyl)-1H-pyrrolo[2,3-b]quinoxaline-3-carboxylic acid (tetrahydro-furan-2-ylmethyl)-amine; 2-amino-1-(3-methoxy-propyl)-1H-pyrrolo[2,3-b]quinoxaline-3-carboxylic acid cyclopentylamide. See, e.g., Nayagam et al. (2006) J. Biolmolec. Screening 11:959.
  • SIRT1 activators include, e.g., stilbene compounds, e.g., ester analogs of resveratrol, e.g., as described in U.S. Patent Publication No. 2008/0255382.
  • suitable SIRT1 activators include, e.g., ester analogs of 3,5,4′-trihydroxy-trans-stilbene.
  • Ester analogs include compounds of the formula:
  • each Y and each Z is independently —O (ethers), —O—C ⁇ O; —C ⁇ O—O (esters); —O—C ⁇ O—O (carbonates); —O—C ⁇ O—NH; —O ⁇ O—NR; —NH—C ⁇ O—O; —NR—C ⁇ O—O (carbamates); —NH—C ⁇ P; —NR—C ⁇ O; —C ⁇ O—NH; —C ⁇ O—NR (primary and secondary amides)-NH; —NR (primary and secondary amines); —N (heterocyclic rings); —S (thiol ethers); and halogen;
  • each n and each m is independently 1, 2, 3, 4, or 5;
  • each A and each B is independently H, R, or absent;
  • each V and each W is independently H, straight or branched alkyl of from 1 to 6 carbon atoms, cycloalkyl of from 3 to 8 carbon atoms, alkoxy, phenyl, benzyl, or halogen,
  • R is an alkyl with at least one carbon atom, an aryl, or an aralkyl.
  • Suitable SIRT1 activators include, e.g., 4′-acetoxy-3,5-bis(methoxymethoxy)stilbene; 4′-acetoxy-3,5-dihydroxystilbene; 3,5-diacetoxy-4′-chloroacetoxy stilbene; 3,5-diacetoxy-4′-hydroxy stilbene; 3,4′-diacetoxy-5-hydroxystilbene; 3-acetoxy-4′5-dihydroxystilbene; and 3,4,5′-triacetoxystilbene.
  • Suitable SIRT1 activators include compounds of any one of Formulas I-VI as described in U.S. Patent Publication No. 2009/0012080.
  • a suitable SIRT1 activator is a compound of the formula:
  • a suitable SIRT1 activator is 4-methyl-N-(2(3-morpholinomethyl)imidazol[2,1-b]thiazol-6-yl)phenyl)-2-(pyridin-3-yl)thiazol-5-carboxamide, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a method of modulating activation and differentiation of a CD4 + T cell, the method generally involving contacting the CD4 + T cell (in vitro, ex vivo, or in vivo) with a SIRT1 inhibitor.
  • the CD4 + T cell is a helper T cell (e.g., a Th1, Th2, or Th17 cell), and the SIRT1 inhibitor inhibits deacetylation of NF- ⁇ B in the CD4 + T cell, thereby increasing CD4 + T helper T cell activity and/or numbers.
  • the CD4 + T cell is a CD4 + /CD25 + /FoxP3 + Treg cell
  • the SIRT1 inhibitor reduces deacetylation of FoxP3 in the CD4 + /CD25 + /FoxP3 + Treg cell, thereby increasing Treg activity and/or numbers.
  • the CD4 + T cell is a helper T cell (e.g., a Th1, a Th2, or a Th17 cell), and the SIRT1 inhibitor inhibits deacetylation of NF- ⁇ B in the CD4 + T cell, thereby increasing CD4 + T helper cell activity and/or numbers.
  • CD4 + T helper cell activity and/or numbers can be increased in vitro, in vivo, or ex vivo.
  • Increasing CD4 + T helper cell activity and/or numbers is useful for increasing an immune response, e.g., increasing an immune response to an antigen.
  • the present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an effective amount of an inhibitor of SIRT1.
  • the method can also be carried out ex vivo, e.g., by (a) contacting CD4 + T helper cells obtained from a donor individual with a SIRT1 inhibitor ex vivo, thereby increasing the activity and/or numbers of CD4 + T helper cells, and (b) returning the CD4 + T helper cells from step (a) to the donor individual or to a recipient individual other than the donor.
  • an effective amount of a SIRT1 inhibitor is an amount that, when contacted with a starting population of CD4 + T helper cells, increases the number of CD4 + T helper cells by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold, or more, compared to the number of CD4 + T helper cells in the starting population before contacting with the SIRT1 inhibitor.
  • the contacting occurs in vitro. In other embodiments, the contacting occurs ex vivo. In other embodiments, the contacting occurs in vivo.
  • an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce deacetylation of NF- ⁇ B in a CD4 + T helper cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of deacetylation of NF- ⁇ B in the CD4 + T helper cell in the absence of the SIRT1 inhibitor.
  • an effective amount of a SIRT1 inhibitor is an amount that, when administered to an individual, increases an immune response in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the immune response in the absence of treatment with the SIRT1 inhibitor.
  • an effective amount of a SIRT1 inhibitor is an amount that increases the number of CD4 + cells in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the number of CD4 + T cells in the individual in the absence of treatment with the SIRT1 inhibitor.
  • an effective amount of a SIRT1 inhibitor is an amount that increases the number of cytotoxic T cells in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the number of cytotoxic T cells in the individual in the absence of treatment with the SIRT1 inhibitor.
  • an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce the number of cancer cells and/or to reduce the volume of a tumor in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the number of cancer cells or compared to the tumor volume in the individual in the absence of treatment with the SIRT1 inhibitor.
  • a SIRT1 inhibitor is administered in conjunction with administration of a tumor-specific antigen.
  • a SIRT1 inhibitor is administered in conjunction with administration of an antigen that induces an immune response to a pathological microorganism.
  • an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce the number of pathological microorganisms in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the number of pathological microorganisms in the individual in the absence of treatment with the SIRT1 inhibitor.
  • a SIRT1 inhibitor is administered in conjunction with administration of an antigen that induces an immune response to a pathological microorganism.
  • an effective amount of a SIRT1 inhibitor is an amount that increases the level of one or more antigen-specific antibodies in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the level of the one or more antigen-specific antibodies in the individual in the absence of treatment with the SIRT1 inhibitor.
  • Analysis can be by any method known in the art, including, but not limited to, measuring antigen-specific antibody production, activation of specific populations of lymphocytes such as CD4 + T cells or NK cells, and/or production of cytokines such as IFN, IL-2, IL-4, or IL-12.
  • Methods for measuring specific antibody responses include enzyme-linked immunosorbent assay (ELISA) and are well known in the art.
  • ELISA enzyme-linked immunosorbent assay
  • Measurement of numbers of specific types of lymphocytes such as CD4 + T cells can be achieved, for example, with fluorescence-activated cell sorting (FACS). Cytotoxicity assays can be performed for instance as described in Raz et al. (1994) Proc. Natl. Acad. Sci.
  • Serum concentrations of cytokines can be measured, for example, by ELISA. These and other assays to evaluate the immune response are well known in the art. In some embodiments, a Th1-type immune response is increased.
  • a subject method of increasing an immune response in an individual is useful for treating various disorders including, e.g., treating an infectious disease (e.g., a disease caused by a pathological microorganism such as a virus, a bacterium, etc.); increasing an immune response to a cancer cell in an individual; enhancing an immune response in an individual who is immunodeficient or immunocompromised; etc.
  • an infectious disease e.g., a disease caused by a pathological microorganism such as a virus, a bacterium, etc.
  • increasing an immune response to a cancer cell in an individual e.g., enhancing an immune response in an individual who is immunodeficient or immunocompromised; etc.
  • a subject method can be used to increase an immune response for treating:
  • viral diseases such as, for example, diseases resulting from infection by an adenovirus, a herpesvirus (e.g., HSV-I, HSV-II, CMV, or VZV), a poxvirus (e.g., an orthopoxvirus such as variola or vaccinia), a picornavirus (e.g., rhinovirus or enterovirus), an orthomyxovirus (e.g., influenzavirus), a paramyxovirus (e.g., parainfluenzavirus, mumps virus, measles virus, and respiratory syncytial virus (RSV)), a coronavirus (e.g., SARS), a papovavirus (e.g., papillomaviruses, such as those that cause genital warts, common warts, or plantar warts), a hepadnavirus (e.g., hepatitis B virus), a flavivirus (e.g., a
  • bacterial diseases such as, for example, diseases resulting from infection by bacteria of, for example, the genus Escherichia, Enterobacter, Salmonella, Staphylococcus, Shigella, Listeria, Aerobacter, Helicobacter, Klebsiella, Proteus, Pseudomonas, Streptococcus, Chlamydia, Mycoplasma, Pneumococcus, Neisseria, Clostridium, Bacillus, Corynebacterium, Mycobacterium, Campylobacter, Vibrio, Serratia, Providencia, Chromobacterium, Brucella, Yersinia, Haemophilus , and Bordetella;
  • (c) other infectious diseases such Chlamydia infection, fungal diseases including but not limited to candidiasis, aspergillosis, histoplasmosis, cryptococcal meningitis, and parasitic diseases including but not limited to malaria, Pneumocystis carinii pneumonia, leishmaniasis, cryptosporidiosis, toxoplasmosis, and trypanosome infection; and
  • neoplastic diseases such as, for example, intraepithelial neoplasias, cervical dysplasia, actinic keratosis, basal cell carcinoma, squamous cell carcinoma, renal cell carcinoma, Kaposi's sarcoma, melanoma, renal cell carcinoma, leukemias including but not limited to myelogeous leukemia, chronic lymphocytic leukemia, multiple myeloma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, B-cell lymphoma, and hairy cell leukemia, and other cancers.
  • leukemias including but not limited to myelogeous leukemia, chronic lymphocytic leukemia, multiple myeloma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, B-cell lymphoma, and hairy cell leukemia, and other cancers.
  • the CD4 + T cell is a CD4 + /CD25 + /FoxP3 + Treg cell
  • the SIRT1 inhibitor reduces deacetylation of FoxP3 in the CD4 + /CD25 + /FoxP3 + Treg cell, thereby increasing Treg activity and/or numbers.
  • the present disclosure provides a method of increasing Treg activity and/or numbers, the method generally involving contacting a na ⁇ ve CD4 + T cell and/or a Treg cell (a CD4 + /CD25 + /FoxP3 + T cell) with a SIRT1 modulator, where the contacting can take place in vitro, ex vivo, or in vivo.
  • the present disclosure provides a method of treating an autoimmune disorder, a method of treating an allergic disorder, a method of reducing graft rejection, and a method of reducing graft-versus-host disease, in an individual, the methods generally involving administering to an individual in need thereof (e.g., an individual having an autoimmune disorder, an individual having an allergic disorder, a graft recipient) an effective amount of an SIRT1 inhibitor.
  • an autoimmune response, an allergic response, graft rejection, or a graft-versus-host response can be reduced by contacting na ⁇ ve CD4 + T cells and/or Treg cells obtained from an individual (e.g., an individual having an autoimmune disorder, an individual having an allergic disorder, a graft recipient) with an SIRT1 inhibitor ex vivo, and returning the Treg cells to the individual.
  • an individual e.g., an individual having an autoimmune disorder, an individual having an allergic disorder, a graft recipient
  • an SIRT1 inhibitor ex vivo
  • an ex vivo method can involve: (a) contacting na ⁇ ve CD4 + T cells and/or Tregs obtained from a donor individual with a SIRT1 inhibitor ex vivo, thereby increasing the activity and/or numbers of Tregs, and (b) returning the Tregs from step (a) to the donor individual or to a recipient individual other than the donor.
  • an effective amount of a SIRT1 inhibitor is an amount that, when contacted with a starting population of Tregs (or a starting population that is a mixture of na ⁇ ve CD4 + T cells and Tregs), increases the number of Tregs by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold, or more, compared to the number of Tregs in the starting population before contacting with the SIRT1 inhibitor.
  • the contacting occurs in vitro. In other embodiments, the contacting occurs ex vivo. In other embodiments, the contacting occurs in vivo.
  • an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce deacetylation of FoxP3 in a Treg by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of deacetylation of FoxP3 in the Treg in the absence of the SIRT1 inhibitor.
  • an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce deacetylation of K31, K263, and K268 (or an equivalent lysine residue) in a FoxP3 polypeptide in a Treg cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of deacetylation of K31, K263, and K268 residues in FoxP3 in the Treg in the absence of the SIRT1 inhibitor.
  • FoxP3 polypeptides are known in the art. Examples of FoxP3 amino acid sequences are presented in FIG. 13 , which shows an alignment of human, mouse, and bovine FoxP3, and which shows the K31, K263, and K268 residues.
  • a FoxP3 polypeptide can comprise an amino acid sequence having at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, with the human FoxP3 amino acid sequence depicted in FIG. 13 .
  • an “effective amount” of a SIRT1 inhibitor is an amount that achieves a degree of immunosuppression sufficient to delay, inhibit, suppress or moderate tissue transplant rejection and/or delay, inhibit, suppress or moderate one or more symptoms of an autoimmune disease and/or delay, inhibit, suppress or moderate an undesired immune response to a foreign antigen such as a therapeutic protein, a viral vector, an allergen, a venom and the like.
  • an “effective amount” of a SIRT1 inhibitor is an amount that reduces an undesired immune response (e.g., an autoimmune response, an allergic response, a graft-versus-host response, a graft rejection) by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the degree of the undesired immune response in the absence of treatment with the SIRT1 inhibitor.
  • an undesired immune response e.g., an autoimmune response, an allergic response, a graft-versus-host response, a graft rejection
  • an SIRT1 inhibitor is effective in reducing an undesired immune response (e.g., an autoimmune response, an allergic response, a graft-versus-host response, a graft rejection) can be determined by measuring one or more well-known parameters associated with the undesired immune response (e.g., the level of autoantibody; the level of auto-reactive T cells; the level of allergen-specific IgE; a symptom of allergy; the level of antibody specific for a graft tissue; etc.).
  • an undesired immune response e.g., an autoimmune response, an allergic response, a graft-versus-host response, a graft rejection
  • parameters associated with the undesired immune response e.g., the level of autoantibody; the level of auto-reactive T cells; the level of allergen-specific IgE; a symptom of allergy; the level of antibody specific for a graft tissue; etc.
  • a subject method is effective to reduce the number and/or activity of an autoreactive cell in an individual by at least about 5%, at least about 10%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%, or more, when compared to the number and/or level of autoreactive cells in the individual not treated with a SIRT1 inhibitor.
  • Whether administration of a SIRT1 inhibitor is effective to reduce the number and/or activity of an autoreactive T lymphocyte in an individual is readily determined using known assays.
  • the number and activity level of autoantigen-specific T lymphocytes is determined using, e.g., a mixed lymphocyte reaction in which irradiated cells comprising a detectable label in the cytoplasm and displaying the autoantigen are mixed with lymphocytes from the individual. Release of detectable label from the cytoplasm of the autoantigen-displaying cells indicates the presence in the individual of autoreactive lymphocytes.
  • Methods of detecting autoreactive T lymphocytes associated with Type 1 diabetes are known in the art; and any such methods can be used. See, e.g., U.S. Pat. No. 6,022,697 for a discussion of a method of detecting autoreactive T lymphocytes associated with Type 1 diabetes.
  • a subject method is useful for treating an autoimmune disease, e.g., for reducing or ameliorating at least one symptom of an autoimmune disease in an individual having an autoimmune disease (e.g., an individual who has been diagnosed as having an autoimmune diseases).
  • Autoimmune diseases include but are not limited to rheumatoid arthritis, Crohn's disease, multiple sclerosis, systemic lupus erythematosus (SLE), autoimmune encephalomyelitis, myasthenia gravis (MG), Hashimoto's thyroiditis, Goodpasture's syndrome, pemphigus (e.g., pemphigus vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis, pernicious anemia, idiopathic Addison's disease, autoimmune-associated infertility, glomerulonephritis (
  • a subject method can be used to increase transplantation tolerance in a subject (e.g., to reduce the likelihood of rejection of the transplant by the transplant recipient).
  • the subject is a recipient of an allogeneic transplant (e.g., the graft is an allograft).
  • Cells, tissues or organs (or parts thereof) that can be transplanted between members of the same species include, but are not limited, to heart, lung, kidney, liver, pancreas, pancreatic islets, brain tissue, cornea, stomach, bone, bone marrow, muscle, intestine, bladder, skin and stem cells.
  • the transplanted tissue or organ is bio-engineered, e.g., when the transplanted tissue or organ is grown from a stem cell or other type of precursor cell(s).
  • Bio-engineered tissue or organ can be grown outside of the body and transplanted directly into the host.
  • the precursor cells or immature organ or tissue is transplanted into the host to grow and mature.
  • the transplanted organ, tissue or cell(s) can also be a xenograft, i.e., the donor is a member of a species different than the recipient.
  • Xenografts are advantageously used with a bio-engineered tissue or organ, which, instead of being transplanted directly into the recipient in need of the tissue or organ, can be transplanted into a surrogate host such as non-human mammal until a suitable human recipient in need of the bio-engineered tissue or organ is identified.
  • the tissue or organ can be transplanted into the surrogate to allow the bio-engineered tissue or organ to mature.
  • Use of surrogate hosts may be preferred in instances where further development of the tissue or organ is required before transplantation into a human recipient.
  • a xenograft is used when a suitable allograft donor is unavailable.
  • a transplanting into a different a species it is desirable to select a host such that the size of the organs in the host and donor are similar.
  • the host is selected to minimize transmission of communicable diseases.
  • a subject method can be used to reduce the incidence and/or severity of an allergic reaction in an individual.
  • a subject method can be used to reduce adverse reaction in an individual to an allergen.
  • Allergens of interest include antigens found in food, such as strawberries, peanuts, milk polypeptides, egg whites, etc.
  • Other allergens of interest include various airborne antigens, such as grass pollens, animal danders, house mite feces, etc.
  • allergens include Dermatophagoides pteryonyssinus (Der P1); Lol pl-V from rye grass pollen; a number of insect venoms, including venom from jumper ant Myrmecia pilosula; Apis mellifera bee venom phospholipase A2 (PLA 2 and antigen 5S; phospholipases from the yellow jacket Vespula maculifrons and white faced hornet Dolichovespula maculata ; a large number of pollen polypeptides, including birch pollen, ragweed pollen, Parol (the major allergen of Parietaria officinalis ) and the cross-reactive allergen Parjl (from Parietaria judaica ), and other atmospheric pollens including Olea europaea, Artemisia sp., gramineae, etc.
  • Dermatophagoides pteryonyssinus Dermatophagoides p
  • allergens of interest are those responsible for allergic dermatitis caused by blood sucking arthropods, e.g. Diptera, including mosquitos ( Anopheles sp., Aedes sp., Culiseta sp., Culex sp.); flies ( Phlebotomus sp., Culicoides sp.) particularly black flies, deer flies and biting midges; ticks ( Dermacenter sp., Ornithodoros sp., Otobius sp.); fleas, e.g. the order Siphonaptera, including the genera Xenopsylla, Pulex and Ctenocephalides felis .
  • Other allergens of interest include drug allergens.
  • SIRT1 inhibitors that are suitable for use in a subject method include, but are not limited to, sirtinol (2-[(2-hydroxynaphthalen-1-ylmethylene)amino]-N-(1-phenyl-ethyl)benzamide); a compound as described in U.S. Pat. No. 7,345,178, provided the compound is an SIRT1 inhibitor, e.g., a compound as shown in Table 5 of U.S. Pat. No. 7,345,178; a compound as described in U.S. Patent Publication No. 2008/0255382; and the like; or a pharmaceutically acceptable salt of any of the foregoing compounds.
  • Suitable SIRT1 inhibitors include, e.g.,
  • Ex-527 is 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide.
  • Ex-527 is a selective SIRT1 inhibitor. See, e.g., Napper et al. (2005) J. Med. Chem. 48:8045.
  • Suitable SIRT1 inhibitors include, e.g., compounds of Table 5 of U.S. Pat. No. 7,345,178, e.g.:
  • Suitable SIRT1 inhibitors include a compound as disclosed in U.S. Patent Publication No. 20008/0214800, or a pharmaceutically acceptable salt of such a compound.
  • suitable SIRT1 inhibitors include, e.g.:
  • Suitable SIRT1 inhibitors include, e.g., 7-chloro-1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; 6-bromo-2,3,4,9-tetrahydro-1H-carbazole-2-carboxylic acid amide; 1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; and 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid (5-chloro-pyridin-2-yl)-amide.
  • a subject treatment method comprises administering to an individual in need thereof an effective amount of a SIRT1 inhibitor or a SIRT1 activator, and at least one additional therapeutic agent.
  • a subject method provides for treating immune hyperactivation that is associated with an immunodeficiency virus infection.
  • a subject method for reducing immunodeficiency virus infection-associated immune hyperactivation involves administering a SIRT1 activator alone, e.g., as monotherapy.
  • a subject method for reducing immunodeficiency virus infection-associated immune hyperactivation involves administering a SIRT1 activator, and at least one additional therapeutic agent, in combination therapy.
  • Suitable additional therapeutic agents include, e.g., therapeutic agents for the treatment of an immunodeficiency virus infection, or for the treatment of a disorder that may accompany an immunodeficiency virus infection (e.g., a bacterial infection, a fungal infection, and the like).
  • a disorder that may accompany an immunodeficiency virus infection e.g., a bacterial infection, a fungal infection, and the like.
  • Suitable additional therapeutic agents include, e.g., beta-lactam antibiotics, tetracyclines, chloramphenicol, neomycin, gramicidin, bacitracin, sulfonamides, nitrofurazone, nalidixic acid, cortisone, hydrocortisone, betamethasone, dexamethasone, fluocortolone, prednisolone, triamcinolone, indomethacin, sulindac, acyclovir, amantadine, rimantadine, recombinant soluble CD4 (rsCD4), anti-receptor antibodies (e.g., for rhinoviruses), nevirapine, cidofovir (VistideTM), trisodium phosphonoformate (FoscarnetTM), famcyclovir, pencyclovir, valacyclovir, nucleic acid/replication inhibitors, interferon, zidovudine (
  • a subject method of increasing an immune response comprises administering to an individual in need thereof a SIRT1 inhibitor and at least one additional therapeutic agent in combined effective amounts to increase an immune response in the individual. In some embodiments, a subject method of increasing an immune response comprises administering to an individual in need thereof a SIRT1 inhibitor and an antigen in combined effective amounts to increase an immune response to the antigen in the individual. In some embodiments, a subject method of increasing an immune response comprises administering to an individual in need thereof a SIRT1 inhibitor and at least one additional therapeutic agent in combined effective amounts to increase an immune response in the individual; and administering an antigen to the individual, where an immune response to the antigen is increased.
  • Suitable additional therapeutic agents include, e.g., immunostimulatory polynucleotides (“ISS”; see, e.g., U.S. Pat. No. 7,479,285); an ISS conjugated to an antigen (see, e.g., U.S. Pat. No. 6,610,661); a Toll-like receptor agonist (see, e.g., U.S. Pat. No. 7,387,271); an immunostimulatory cytokine (e.g., IL-2); and the like.
  • ISS immunostimulatory polynucleotides
  • ISS immunostimulatory polynucleotides
  • an antigen see, e.g., U.S. Pat. No. 6,610,661
  • Toll-like receptor agonist see, e.g., U.S. Pat. No. 7,387,271
  • an immunostimulatory cytokine e.g., IL-2
  • Suitable ISS include poly
  • ISS can be from about 6 nucleotides in length to about 200 nucleotides in length, or longer than 200 nucleotides in length.
  • An ISS can comprise a phosphate backbone modification, where backbone phosphate group modifications include, e.g., methylphosphonate, phosphorothioate, phosphoroamidate and phosphorodithioate internucleotide linkages.
  • TLR agonists include, e.g., TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8 and TLR9 agonists.
  • Suitable TLR agonists include, e.g., an imidazoquinoline amine, a tetrahydroimidazoquinoline amine, an imidazopyridine amine, a 1,2-bridged imidazoquinoline amine, a 6,7-fused cycloalkylimidazopyridine amine, an imidazonaphthyridine amine, a tetrahydroimidazonaphthyridine amine, an oxazoloquinoline amine, a thiazoloquinoline amine, an oxazolopyridine amine, a thiazolopyridine amine, an oxazolonaphthyridine amine, and a thiazolonaphthyridine amine.
  • a subject method of reducing an undesired immune response involves administering a SIRT1 inhibitor alone, e.g., as monotherapy.
  • a subject method of reducing an undesired immune response involves administering a SIRT1 inhibitor in combination therapy with one or more additional therapeutic agents.
  • a SIRT1 inhibitor can be administered in combination therapy with one or more agents for treating an autoimmune disorder.
  • agents that are suitable for treating Type 1 diabetes include insulin, including naturally occurring insulin, insulin analogs, and the like.
  • Interferon-alpha is an agent that is currently in use for treating multiple sclerosis, and can be used in combination therapy with a SIRT1 inhibitor.
  • Other agents that are currently in use for treating autoimmune disorders include corticosteroid drugs; non-steroidal anti-inflammatory drugs (NSAIDs); and immunosuppressant drugs such as cyclophosphamide, methotrexate, and azathioprine.
  • a SIRT1 inhibitor can be administered in combination therapy with one or more agents for treating an allergic disorder.
  • agents include, but are not limited to, antihistamines such as diphenhydramine (Benadryl); epinephrine; decongestants; and the like.
  • a SIRT1 inhibitor can be administered in combination therapy with one or more agents for reducing graft rejection or GVHD.
  • Agents suitable for reducing graft rejection or GVHD include, but are not limited to, glucocorticoids (e.g., cortisol; dexamethasone; etc.); cytostatic agents (e.g., cyclophosphamide; nitrosoureas; platinum compounds; methotrexate; azathioprine; mercaptopurine; dactinomycin; anthracyclines; mitomycin C; bleomycin; mitramycin; etc.); an anti-CD20 monoclonal antibody; cyclosporin; tacrolimus; voclosporin; sirolimus; and the like.
  • glucocorticoids e.g., cortisol; dexamethasone; etc.
  • cytostatic agents e.g., cyclophosphamide; nitrosourea
  • a SIRT1 activator or a SIRT1 inhibitor can be formulated with one or more pharmaceutically acceptable excipients.
  • SIRT1 activators and SIRT1 inhibitors are referred to collectively below as “active agent” or “drug.”
  • active agent or “drug.”
  • a wide variety of pharmaceutically acceptable excipients are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, A. Gennaro (2000) “Remington: The Science and Practice of Pharmacy,” 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H. C.
  • the pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are readily available to the public.
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of a subject active agent calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for an active agent depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • a SIRT1 activator or a SIRT1 inhibitor may be administered to the host using any convenient means capable of resulting in the desired outcome, e.g., reduction of disease, reduction of a symptom of a disease, etc.
  • a SIRT1 activator or a SIRT1 inhibitor can be incorporated into a variety of formulations for therapeutic administration.
  • a SIRT1 activator or a SIRT1 inhibitor can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof.
  • the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985.
  • the composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the subject being treated.
  • a SIRT1 activator or a SIRT1 inhibitor may be administered in the form of its pharmaceutically acceptable salts, or an active agent may be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • active agent may be administered in the form of its pharmaceutically acceptable salts, or an active agent may be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • an active agent for oral preparations, an active agent (a SIRT1 activator or a SIRT1 inhibitor) can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxy
  • An active agent can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • an aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol
  • solubilizers such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • An active agent can be utilized in aerosol formulation to be administered via inhalation.
  • An active agent can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.
  • an active agent can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • An active agent can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycol monomethyl ethers, which melt at body temperature, yet are solidified at room temperature.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the subject active agent.
  • unit dosage forms for injection or intravenous administration may comprise a subject active agent in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • An active agent can be formulated for administration by injection.
  • injectable compositions are prepared as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared.
  • the preparation may also be emulsified or the active ingredient encapsulated in liposome vehicles.
  • an active agent is delivered by a continuous delivery system.
  • continuous delivery system is used interchangeably herein with “controlled delivery system” and encompasses continuous (e.g., controlled) delivery devices (e.g., pumps) in combination with catheters, injection devices, and the like, a wide variety of which are known in the art.
  • Mechanical or electromechanical infusion pumps can also be suitable for use with the present invention.
  • Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852; 5,820,589; 5,643,207; 6,198,966; and the like.
  • delivery of active agent can be accomplished using any of a variety of refillable, pump systems. Pumps provide consistent, controlled release over time.
  • the active agent is in a liquid formulation in a drug-impermeable reservoir, and is delivered in a continuous fashion to the individual.
  • the drug delivery system is an at least partially implantable device.
  • the implantable device can be implanted at any suitable implantation site using methods and devices well known in the art.
  • An implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned.
  • Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body. Subcutaneous implantation sites are used in some embodiments because of convenience in implantation and removal of the drug delivery device.
  • Drug release devices suitable for use in the invention may be based on any of a variety of modes of operation.
  • the drug release device can be based upon a diffusive system, a convective system, or an erodible system (e.g., an erosion-based system).
  • the drug release device can be an electrochemical pump, osmotic pump, an electroosmotic pump, a vapor pressure pump, or osmotic bursting matrix, e.g., where the drug (“active agent”) is incorporated into a polymer and the polymer provides for release of drug formulation concomitant with degradation of a drug-impregnated polymeric material (e.g., a biodegradable, drug-impregnated polymeric material).
  • the drug release device is based upon an electrodiffusion system, an electrolytic pump, an effervescent pump, a piezoelectric pump, a hydrolytic system, etc.
  • Drug release devices based upon a mechanical or electromechanical infusion pump can also be suitable for use with the present invention.
  • Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852, and the like.
  • a subject treatment method can be accomplished using any of a variety of refillable, non-exchangeable pump systems. Pumps and other convective systems are generally preferred due to their generally more consistent, controlled release over time. Osmotic pumps are used in some embodiments due to their combined advantages of more consistent controlled release and relatively small size (see, e.g., PCT published application no. WO 97/27840 and U.S. Pat. Nos.
  • Exemplary osmotically-driven devices suitable for use in the invention include, but are not necessarily limited to, those described in U.S. Pat. Nos. 3,760,984; 3,845,770; 3,916,899; 3,923,426; 3,987,790; 3,995,631; 3,916,899; 4,016,880; 4,036,228; 4,111,202; 4,111,203; 4,203,440; 4,203,442; 4,210,139; 4,327,725; 4,627,850; 4,865,845; 5,057,318; 5,059,423; 5,112,614; 5,137,727; 5,234,692; 5,234,693; 5,728,396; and the like.
  • the drug delivery device is an implantable device.
  • the drug delivery device can be implanted at any suitable implantation site using methods and devices well known in the art.
  • an implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned. Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body.
  • an active agent (a SIRT1 activator or a SIRT1 inhibitor) is delivered using an implantable drug delivery system, e.g., a system that is programmable to provide for administration of an active agent.
  • implantable drug delivery system e.g., a system that is programmable to provide for administration of an active agent.
  • implantable drug delivery system e.g., a system that is programmable to provide for administration of an active agent.
  • implantable drug delivery system e.g., a system that is programmable to provide for administration of an active agent.
  • Exemplary programmable, implantable systems include implantable infusion pumps.
  • Exemplary implantable infusion pumps, or devices useful in connection with such pumps, are described in, for example, U.S. Pat. Nos. 4,350,155; 5,443,450; 5,814,019; 5,976,109; 6,017,328; 6,171,276; 6,241,704; 6,464,687; 6,475,180; and 6,512,954.
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof.
  • the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985.
  • the composition or formulation to be administered will, in any event, contain a quantity of an active agent adequate to achieve the desired state in the subject being treated.
  • an active agent (a SIRT1 activator or a SIRT1 inhibitor) is formulated for oral delivery to an individual in need of such an agent.
  • a formulation comprising an active agent will in some embodiments include an enteric-soluble coating material.
  • Suitable enteric-soluble coating material include hydroxypropyl methylcellulose acetate succinate (HPMCAS), hydroxypropyl methyl cellulose phthalate (HPMCP), cellulose acetate phthalate (CAP), polyvinyl phthalic acetate (PVPA), EudragitTM, and shellac.
  • an active agent is formulated with one or more pharmaceutical excipients and coated with an enteric coating, as described in U.S. Pat. No. 6,346,269.
  • a solution comprising an active agent and a stabilizer is coated onto a core comprising pharmaceutically acceptable excipients, to form an active agent-coated core; a sub-coating layer is applied to the active agent-coated core, which is then coated with an enteric coating layer.
  • the core generally includes pharmaceutically inactive components such as lactose, a starch, mannitol, sodium carboxymethyl cellulose, sodium starch glycolate, sodium chloride, potassium chloride, pigments, salts of alginic acid, talc, titanium dioxide, stearic acid, stearate, micro-crystalline cellulose, glycerin, polyethylene glycol, triethyl citrate, tributyl citrate, propanyl triacetate, dibasic calcium phosphate, tribasic sodium phosphate, calcium sulfate, cyclodextrin, and castor oil.
  • Suitable solvents for an active agent include aqueous solvents.
  • Suitable stabilizers include alkali-metals and alkaline earth metals, bases of phosphates and organic acid salts and organic amines.
  • the sub-coating layer comprises one or more of an adhesive, a plasticizer, and an anti-tackiness agent.
  • Suitable anti-tackiness agents include talc, stearic acid, stearate, sodium stearyl fumarate, glyceryl behenate, kaolin and aerosil.
  • Suitable adhesives include polyvinyl pyrrolidone (PVP), gelatin, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), vinyl acetate (VA), polyvinyl alcohol (PVA), methyl cellulose (MC), ethyl cellulose (EC), hydroxypropyl methyl cellulose phthalate (HPMCP), cellulose acetate phthalates (CAP), xanthan gum, alginic acid, salts of alginic acid, EudragitTM copolymer of methyl acrylic acid/methyl methacrylate with polyvinyl acetate phthalate (PVAP).
  • PVAP polyvinyl pyrrolidone
  • gelatin gelatin
  • HEC hydroxyethyl cellulose
  • HPC hydroxypropyl cellulose
  • HPMC hydroxypropyl methyl cellulose
  • VA vinyl acetate
  • PVA polyvinyl alcohol
  • MC methyl
  • Suitable plasticizers include glycerin, polyethylene glycol, triethyl citrate, tributyl citrate, propanyl triacetate and castor oil.
  • Suitable enteric-soluble coating material include hydroxypropyl methylcellulose acetate succinate (HPMCAS), hydroxypropyl methyl cellulose phthalate (HPMCP), cellulose acetate phthalate (CAP), polyvinyl phthalic acetate (PVPA), EudragitTM and shellac.
  • Suitable oral formulations also include an active agent formulated with any of the following: microgranules (see, e.g., U.S. Pat. No. 6,458,398); biodegradable macromers (see, e.g., U.S. Pat. No. 6,703,037); biodegradable hydrogels (see, e.g., Graham and McNeill (1989) Biomaterials 5:27-36); biodegradable particulate vectors (see, e.g., U.S. Pat. No. 5,736,371); bioabsorbable lactone polymers (see, e.g., U.S. Pat. No. 5,631,015); slow release protein polymers (see, e.g., U.S. Pat.
  • Suitable oral formulations also include an active agent with any of the following: a carrier such as Emisphere® (Emisphere Technologies, Inc.); TIMERx, a hydrophilic matrix combining xanthan and locust bean gums which, in the presence of dextrose, form a strong binder gel in water (Penwest); GeminexTM (Penwest); ProciseTM (GlaxoSmithKline); SAVITTM (Mistral Pharma Inc.); RingCapTM (Alza Corp.); Smartrix® (Smartrix Technologies, Inc.); SQZgelTM (MacroMed, Inc.); GeomatrixTM (Skye Pharma, Inc.); Oros® Tri-layer (Alza Corporation); and the like.
  • a carrier such as Emisphere® (Emisphere Technologies, Inc.); TIMERx, a hydrophilic matrix combining xanthan and locust bean gums which, in the presence of dextrose, form a strong binder gel in water (Pen
  • formulations such as those described in U.S. Pat. No. 6,296,842 (Alkermes Controlled Therapeutics, Inc.); U.S. Pat. No. 6,187,330 (Scios, Inc.); and the like.
  • Suitable intestinal absorption enhancers include, but are not limited to, calcium chelators (e.g., citrate, ethylenediamine tetracetic acid); surfactants (e.g., sodium dodecyl sulfate, bile salts, palmitoylcarnitine, and sodium salts of fatty acids); toxins (e.g., zonula occludens toxin); and the like.
  • calcium chelators e.g., citrate, ethylenediamine tetracetic acid
  • surfactants e.g., sodium dodecyl sulfate, bile salts, palmitoylcarnitine, and sodium salts of fatty acids
  • toxins e.g., zonula occludens toxin
  • An active agent (a SIRT1 activator or a SIRT1 inhibitor) will in some embodiments be administered to a patient by means of a pharmaceutical delivery system for the inhalation route.
  • An active agent can be formulated in a form suitable for administration by inhalation.
  • the inhalational route of administration provides the advantage that the inhaled drug can bypass the blood-brain barrier.
  • the pharmaceutical delivery system is one that is suitable for respiratory therapy by delivery of an active agent to mucosal linings of the bronchi.
  • This invention can utilize a system that depends on the power of a compressed gas to expel the active agent from a container.
  • An aerosol or pressurized package can be employed for this purpose.
  • the term “aerosol” is used in its conventional sense as referring to very fine liquid or solid particles carries by a propellant gas under pressure to a site of therapeutic application.
  • the aerosol contains an active agent, which can be dissolved, suspended, or emulsified in a mixture of a fluid carrier and a propellant.
  • the aerosol can be in the form of a solution, suspension, emulsion, powder, or semi-solid preparation. Aerosols employed in the present invention are intended for administration as fine, solid particles or as liquid mists via the respiratory tract of a patient.
  • propellants include, but are not limited to, hydrocarbons or other suitable gas.
  • the dosage unit may be determined by providing a value to deliver a metered amount.
  • An active agent can also be formulated for delivery with a nebulizer, which is an instrument that generates very fine liquid particles of substantially uniform size in a gas.
  • a liquid containing the active agent is dispersed as droplets.
  • the small droplets can be carried by a current of air through an outlet tube of the nebulizer. The resulting mist penetrates into the respiratory tract of the patient.
  • a powder composition containing an active agent, with or without a lubricant, carrier, or propellant, can be administered to a mammal in need of therapy.
  • This embodiment of the invention can be carried out with a conventional device for administering a powder pharmaceutical composition by inhalation.
  • a powder mixture of the compound and a suitable powder base such as lactose or starch may be presented in unit dosage form in for example capsular or cartridges, e.g. gelatin, or blister packs, from which the powder may be administered with the aid of an inhaler.
  • an active agent can be formulated in basically three different types of formulations for inhalation.
  • an active agent can be formulated with low boiling point propellants.
  • Such formulations are generally administered by conventional meter dose inhalers (MDI's).
  • MDI's can be modified so as to increase the ability to obtain repeatable dosing by utilizing technology which measures the inspiratory volume and flow rate of the patient as discussed within U.S. Pat. Nos. 5,404,871 and 5,542,410.
  • an active agent can be formulated in aqueous or ethanolic solutions and delivered by conventional nebulizers.
  • solution formulations are aerosolized using devices and systems such as disclosed within U.S. Pat. Nos. 5,497,763; 5,544,646; 5,718,222; and 5,660,166.
  • An active agent can be formulated into dry powder formulations. Such formulations can be administered by simply inhaling the dry powder formulation after creating an aerosol mist of the powder. Technology for carrying such out is described within U.S. Pat. No. 5,775,320 and U.S. Pat. No. 5,740,794.
  • an active agent can be administered in dosages of, for example, 0.1 ⁇ g to 500 mg/kg body weight per day, e.g., from about 0.1 ⁇ g/kg body weight per day to about 1 ⁇ g/kg body weight per day, from about 1 ⁇ g/kg body weight per day to about 25 ⁇ g/kg body weight per day, from about 25 ⁇ g/kg body weight per day to about 50 ⁇ g/kg body weight per day, from about 50 ⁇ g/kg body weight per day to about 100 ⁇ g/kg body weight per day, from about 100 ⁇ g/kg body weight per day to about 500 ⁇ g/kg body weight per day, from about 500 ⁇ g/kg body weight per day to about 1 mg/kg body weight per day, from about 1 mg/kg body weight per day to about 25 mg/kg body weight per day, from about 25 mg/kg body weight per day to about 50 mg/kg body weight per day, from about 50 mg/kg body weight per day to about 100 mg
  • oral dosages may be about ten times the injection dose. Higher doses may be used for localized routes of delivery.
  • an active agent can be administered in an amount of from about 1 mg to about 1000 mg per dose, e.g., from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 50 mg, from about 50 mg to about 75 mg, from about 75 mg to about 100 mg, from about 100 mg to about 125 mg, from about 125 mg to about 150 mg, from about 150 mg to about 175 mg, from about 175 mg to about 200 mg, from about 200 mg to about 225 mg, from about 225 mg to about 250 mg, from about 250 mg to about 300 mg, from about 300 mg to about 350 mg, from about 350 mg to about 400 mg, from about 400 mg to about 450 mg, from about 450 mg to about 500 mg, from about 500 mg to about 750 mg, or from about 750 mg to about 1000 mg per dose.
  • An exemplary dosage may be a solution suitable for intravenous administration; a tablet taken from two to six times daily, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active agent, etc.
  • the time-release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release.
  • dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects.
  • Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means.
  • a suitable dosage range is in some embodiments one which provides up to about 1 ⁇ g to about 1,000 ⁇ g or about 10,000 ⁇ g of active agent in a blood sample taken from the individual being treated, about 24 hours after administration of the active agent to the individual.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more active agents.
  • unit dosage forms for injection or intravenous administration may comprise the active agent(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • multiple doses of an active agent are administered.
  • the frequency of administration of an active agent can vary depending on any of a variety of factors, e.g., severity of the symptoms, etc.
  • an active agent is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid).
  • an active agent is administered continuously.
  • an active agent can vary, depending on any of a variety of factors, e.g., patient response, etc.
  • an active agent can be administered over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
  • an active agent is administered for the lifetime of the individual.
  • An active agent is administered to an individual using any available method and route suitable for drug delivery, including in vivo and ex vivo methods, as well as systemic and localized routes of administration.
  • Administration can be acute (e.g., of short duration, e.g., a single administration, administration for one day to one week), or chronic (e.g., of long duration, e.g., administration for longer than one week, e.g., administration over a period of time of from about 2 weeks to about one month, from about one month to about 3 months, from about 3 months to about 6 months, from about 6 months to about 1 year, or longer than one year).
  • routes of administration include intranasal, intramuscular, intratracheal, subcutaneous, intradermal, transdermal, sublingual, topical application, intravenous, ocular (e.g., topically to the eye, intravitreal, etc.), rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration may be combined, if desired, or adjusted depending upon the agent and/or the desired effect.
  • the active agent can be administered in a single dose or in multiple doses.
  • An active agent can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes.
  • routes of administration contemplated by the invention include, but are not necessarily limited to, enteral, parenteral, and inhalational routes.
  • Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular, intraorbital, intracapsular, intraspinal, intrasternal, ocular, and intravenous routes, i.e., any route of administration other than through the alimentary canal.
  • Parenteral administration can be carried to effect systemic or local delivery of the agent. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
  • An active agent can also be delivered to the subject by enteral administration.
  • Enteral routes of administration include, but are not necessarily limited to, oral and rectal (e.g., using a suppository) delivery.
  • Methods of administration of an active agent through the skin or mucosa include, but are not necessarily limited to, topical application of a suitable pharmaceutical preparation, transdermal transmission, injection and epidermal administration.
  • a suitable pharmaceutical preparation for transdermal transmission, absorption promoters or iontophoresis are suitable methods.
  • Iontophoretic transmission may be accomplished using commercially available “patches” which deliver their product continuously via electric pulses through unbroken skin for periods of several days or more.
  • Subjects suitable for treatment with a subject method include individuals in need of such treatment.
  • Subjects suitable for treatment with a subject method of reducing chronic immune hyperactivity include individuals who have chronic immune hyperactivity due to an infection with an immunodeficiency virus, e.g., human immunodeficiency virus-1, human immunodeficiency virus-2.
  • an immunodeficiency virus e.g., human immunodeficiency virus-1, human immunodeficiency virus-2.
  • Subjects suitable for treatment a subject method of increasing an immune response include an individual who has been infected with a pathogenic microorganism; an individual who is susceptible to infection by a pathogenic microorganism, but who has not yet been infected; and an individual who has cancer.
  • Subjects suitable for treatment with a subject method of increasing an immune response include pediatric target populations, e.g., individuals between about 1 year of age and about 17 years of age, including infants (e.g., from about 1 month old to about 1 year old); children (e.g., from about 1 year old to about 12 years old); and adolescents (e.g., from about 13 years old to about 17 years old).
  • pediatric target populations e.g., individuals between about 1 year of age and about 17 years of age, including infants (e.g., from about 1 month old to about 1 year old); children (e.g., from about 1 year old to about 12 years old); and adolescents (e.g., from about 13 years old to about 17 years old).
  • Subjects suitable for treatment with a subject method of increasing an immune response include adult individuals.
  • Subjects suitable for treatment with a subject method of increasing an immune response include immunodeficient individuals, e.g., individuals with an acquired immunodeficiency, e.g., as a results of radiation therapy for cancer, as a result of corticosteroid treatment, as a result of cancer chemotherapy, etc. Also suitable for treatment with a subject method of increasing an immune response are individuals who have undergone bone marrow transplantation or any other organ transplantation, e.g., immunosuppressed individuals.
  • Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pl, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal(ly); s.c., subcutaneous(ly); and the like.
  • “a” preceding a protein denotes an antibody to that protein; for example, ⁇ -p65 is an antibody to p65.
  • HeLa, 293, 293T and Jurkat cells were cultured under standard tissue culture conditions. MEF cells derived from SIRT1 ⁇ / ⁇ cells were grown as described (Pagans et al., 2005). Mutant constructs for SIRT1 HDAC domain were prepared by site-directed mutagenesis using FLAG-tagged SIRT1 as a template.
  • the Sir2 ⁇ deletion constructs were kindly provided by V. Sartorelli, NIH, the 3 ⁇ - ⁇ B luciferase reporter by N. Chirmule, Merck Inc, the I ⁇ Ba luciferase reporter by K. Yamamoto, UCSF, and the E-selectin luciferase reporter by J. Pober, Yale University.
  • LTR-GFP and LTR-Tat-GFP are HIV-based vectors derived from the pHR′ series, in which GFP alone or Tat (101 aa) and GFP are under the control of the HIV-1 LTR through the use of an internal ribosomal entry site (Jordan et al., 2001).
  • the EF-1 ⁇ promoter was inserted into the LTR-GFP vector upstream of wild type or mutant Tat (EF1 ⁇ -Tat-GFP).
  • Each vector was cotransfected into 293T cells together with a packaging construct (pCMV-R8.91) that provides all HIV genes required for production of infective particles and a plasmid encoding the vesicular stomatitis virus envelope G protein (VSV-G) to produce pseudotyped viral particles with broad host range and high infectivity.
  • Viral supernatant containing 1500 ng of p24 was used to infect 18 ⁇ 10 6 Jurkat T cells. Infections were carried out in 6-well plates at 2400 rpm in a Beckman-Coulter centrifuge for 2 h at 32° C. in the presence of polybrene (1 ⁇ g/ml, Sigma).
  • Flow cytometry analysis of GFP expression (FACSCalibur, BD Bioscience) showed that infection efficiencies ranged from 70-98% GFP cells in individual experiments. Tat expression was visualized by western blotting with polyclonal ⁇ -FLAG and ⁇ - ⁇ -actin (Sigma) antibodies.
  • Open reading frames corresponding to Myc-tagged human SIRT1 and T7-tagged HIV-1 Tat (101 aa) were inserted into the murine stem cell virus (MSCV)-based retroviral vectors MSCV-puromycin and MSCV-zeocin, respectively (Clontech).
  • MSCV murine stem cell virus
  • To obtain recombinant virus 10 ⁇ g of each construct or empty control vectors were transfected into BOSC23 cells, a retroviral packaging cell line derived from 293 cells. The supernatants were collected 48 h after transfection and filtered through a 0.45- ⁇ m membrane.
  • SIRT1 ⁇ / ⁇ MEF cells (2 ⁇ 10 5 ) were incubated with 2 ml of the supernatant containing SIRT1-expressing retrovirus or control virus together with polybrene (8 ⁇ g/ml, Sigma). Cells were selected after 48 h with puromycin (2.5 ⁇ g/ml, Invitrogen). Polyclonal puromycin-resistant MEF cells were reinfected with Tat-expressing or empty control retroviruses and selected in the presence of both puromycin (2.5 ⁇ g/ml) and zeocin (100 ⁇ g/ml, Invitrogen).
  • RNA was extracted using RNA STAT-60 reagent (Tel-Test) according to the manufacturer's instruction. The first strand cDNA was generated using 2 ⁇ g of total RNA and SuperScript reverse transcriptase (Invitrogen). Human IL-2 and murine E-selectin mRNAs were quantified by QuantiTect gene expression assays (Qiagen). Murine I ⁇ B ⁇ and murine/human GAPDH mRNA concentrations were determined with SYBR Green I master mix (MCLab). Relative gene expression ratios between stimulated and nonstimulated conditions were calculated by the equation, ratio 2 ⁇ Ct , comparative Ct method Applied Biosystems).
  • Luciferase reporter constructs and protein expression vectors were transiently cotransfected into HeLa cells using Lipofectamine (Invitrogen). 24 h after transfection, cells were lysed and processed for luciferase assays (Promega). P values (paired t-test) were used for statistical analysis.
  • Full-length human SIRT1 (His-tagged) was purified using Ni-NTA agarose (Qiagen) and Superdex 200 gel filtration. The rate of 1 ⁇ M SIRT1 was calculated for 200 ⁇ M fluorogenic deacetylase substrate (BioMol) in the presence of twelve concentrations (1, 2, 3, 4, 5, 6, 10, 15, 25, 50, 126, 252 ⁇ M) of synthetic Tat (72 aa; Peptide Specialty Laboratory, Heidelberg, Germany).
  • Expression plasmids for FLAG-tagged wild type and mutants SIRT1 (1 ⁇ g) were transfected in 293 cells with lipofectamine reagent. Equal amounts of whole cell extracts (2 mg) were immunoprecipitated and in vitro HDAC assays were performed as previously described (Pagans et al., 2005).
  • Recombinant p65 protein prepared from baculovirus-infected Sf9 insect cells was acetylated by immunoprecipitated p300 overexpressed in 293 cells as previously described (Chen et al., 2005). Acetylated p65 protein was incubated with recombinant SIRT1 (1 ⁇ g/5 U; Biomol) in SIRT1 deacetylase buffer (50 mM Tris-HCl pH 9, 4 mM MgCl 2 , 0.2 mM DTT) in the presence of NAD (1 mM; Sigma) for 3 h at 37° C.
  • Reactions containing synthetic Tat (0.1 ⁇ g) or nicotinamide (10 mM) were preincubated for 15 mM at room temperature. Reactions were stopped by the addition of SDS loading buffer, boiled, and after brief centrifugation, analyzed by western blotting with rabbit ⁇ -AcK310 p65 (Chen et al., 2005) or mouse ⁇ -p65 antibodies (sc-8008, Santa Cruz).
  • Tat To recapitulate the effect of Tat on immune activation in a manner as close to natural HIV infection as possible, we employed an HIV-based lentiviral vector in which both FLAG-tagged Tat and green fluorescent protein (GFP) are expressed under the control of the HIV promoter in the 5′-LTR.
  • Jurkat T cells were infected with viral particles containing this vector or a control vector expressing only GFP. Infected cultures were stimulated with antibodies specific for the CD3 and CD28 receptors to mimic physiological T-cell activation.
  • Tat expression was visualized by western blot analysis ( FIG. 1A ).
  • IL-2 expression in control cells was only induced ⁇ 250 fold ( FIG. 1B ).
  • FIG. 1C A similar difference was observed when IL-2 protein was measured in the culture supernatant ( FIG. 1C ). No effect of Tat was observed in nonactivated cultures.
  • FIGS. 1A-F Tat and nicotinamide cause T-cell hyperactivation
  • A Western blot analysis of Tat protein in Jurkat T cells infected with lentiviral vectors expressing LTR-GFP and LTR-Tat-GFP. Infected cultures were activated for 2 h and 8 h with ⁇ -CD3/28 antibodies.
  • B Real-time RT-PCR analysis of IL-2 mRNA levels in infected cultures 2 h after activation with ⁇ -CD3/28. Results are expressed as fold induction by ⁇ -CD3/28 treatment.
  • C IL-2 protein levels measured by ELISA in the supernatant of infected cultures 8 h after ⁇ -CD3/28 treatment.
  • E Real-time RT-PCR analysis of IL-2 mRNA levels in Jurkat T cells infected with lentiviral vectors after treatment with nicotinamide and activation with ⁇ -CD3/28 antibodies.
  • F Western blot analysis of Tat protein in Jurkat T cells infected with lentiviral vectors after treatment with nicotinamide and activation with ⁇ -CD3/28. In B-E averages of three independent experiments ( ⁇ SEM) are shown.
  • SIRT1 is Required for the Hyperactivation of NF- ⁇ B Target Genes by Tat
  • T-cell activation is coupled to the stimulation of several transcription factors that regulate the transcriptional status of the IL-2 gene.
  • Efforts were focused on NF- ⁇ B since its activity is regulated by SIRT1 (Yeung et al., 2004).
  • Tat enhanced IL-2 promoter activity via the NF- ⁇ B binding sites present within the IL-2 promoter (Fortin et al., 2004; Ott et al., 1997; Ott et al., 1998; Westendorp et al., 1994).
  • Tat like nicotinamide, inhibits the SIRT1 deacetylase activity leading to hyperactivation of NF- ⁇ B-responsive genes including IL-2.
  • E-selectin another NF- ⁇ B-responsive gene that is also superinduced by Tat (Cota-Gomez et al., 2002; Lee et al., 2004).
  • TNF ⁇ treatment endogenous E-selectin gene expression was enhanced 6-fold by Tat in SIRT1-expressing, but not in SIRT1-negative cells ( FIG. 2C ).
  • the Tat effect was only observed at 2 h after TNF ⁇ treatment.
  • no induction of E-selectin gene expression was evident, consistent with the prior finding that E-selectin mRNA accumulation peaks 2 h after TNF ⁇ treatment and then rapidly declines (Edelstein et al., 2005).
  • Tat had no effect on the constitutive expression of the endogenous glyceraldehyde-3-phosphate dehydrogenase gene (GAPDH) ( FIG. 2D ).
  • FIGS. 2A-D Tat-mediated superinduction of NF- ⁇ B-responsive genes requires SIRT1
  • A Western blot analysis of SIRT1, Tat and ⁇ -actin in SIRT1 ⁇ / ⁇ MEF cells infected with MSCV-based retroviral particles expressing SIRT1 or Tat.
  • B-D Real-time RT-PCR analysis of I ⁇ B ⁇ (B), E-selectin (C), and GAPDH (D) mRNA levels in the four polyclonal MEF populations described in (A) after incubation with TNF ⁇ (20 ng/ml) for 2 h and 8 h. Data are presented relative to values obtained in cells lacking Tat (100%). The mean of three independent experiments ( ⁇ SEM) is shown.
  • SIRT1 No change in expression levels of p65 was observed by western blotting in the presence of SIRT1 or Tat.
  • the suppressive activity of SIRT1 was dependent on its intrinsic deacetylase activity of SIRT1, since a catalytically inactive SIRT1 mutant (P447E; shown in FIG. 4D ) did not suppress the activity of p65 and was unresponsive to the action of Tat.
  • Expression of Tat did not affect basal or p65-mediated activity of the I ⁇ B ⁇ promoter in the absence of SIRT1 expression ( FIG. 3A ).
  • FIGS. 3A-C Tat neutralizes the inhibitory effect of SIRT1 on NF- ⁇ B-responsive promoters.
  • Promoter reporter assays using (A) the I ⁇ B ⁇ , (B) the E-selectin, and (C) the 3 ⁇ - ⁇ B luciferase reporter construct.
  • Luciferase constructs (0.2 ⁇ g) were transiently cotransfected with expression vectors for p65 or p65 K310R (1 ng), SIRT1 or SIRT1 P447E (0.2 ⁇ g), and Tat (50 ng) into HeLa cells. The mean of three independent experiments ( ⁇ SEM) is shown.
  • the region spanning amino acids 341-512 contains the catalytic domain of SIRT1, which is shared among all sirtuins. It is 98% conserved in the murine and human SIRT1 proteins and consists of two distinct domains: a large Rossmann fold domain characteristic of NAD + -binding proteins and a smaller domain containing a structural zinc ion. The NAD and acetyl-lysine substrate bind in the active site cleft between the two domains (Avalos et al., 2002).
  • FIGS. 4A-E Tat binds to the acetyl lysine-binding site in SIRT1
  • A Communoprecipitation assay of Tat and SIRT1 deletion mutants. Expression vectors for Myc-tagged murine Sir2 ⁇ deletion mutants or full length human SIRT1 (each 1 ⁇ g) were transiently cotransfected with constructs expressing FLAG-tagged Tat (1 ⁇ g) in 293 cells. Immunoprecipitations were performed with ⁇ -FLAG agarose and western blotting with ⁇ -Myc and ⁇ -FLAG antibodies.
  • B Schematic summary of Tat binding to murine Sir2 ⁇ deletion mutants. The relative localization of the human SIRT1 HDAC domain is shown at the bottom.
  • FIG. 1 Schematic representation of the human SIRT1 HDAC domain with point mutations.
  • D In vitro HDAC assays of immunoprecipitated human SIRT1 mutants with and without NAD + .
  • E Communoprecipitation of SIRT1 mutants and Tat in 293 cells. Cell extracts were immunoprecipitated with ⁇ -T7 agarose followed by western blotting with ⁇ -FLAG and ⁇ -T7 antibodies.
  • Tat Inhibits the SIRT1 Deacetylase Activity and Induces Hyperacetylation of NF- ⁇ B
  • FIGS. 5A-D Tat inhibits the SIRT1 deacetylase activity
  • A In vitro deacetylation assay of recombinant SIRT1 in the presence of synthetic Tat (aa 1-72). The rate of 1 ⁇ M SIRT1 was calculated for 200 ⁇ M fluorogenic deacetylase substrate (aa 379-382 of p53) in the presence of twelve concentrations of synthetic Tat. The data were plotted in Prism with X equal to Log 10 [Tat] and Y equal to rate. A nonlinear regression was done using the one-site competitive binding equation to determine the IC 50 .
  • Total cell lysates were analyzed by western blotting with ⁇ -AcK310 p65 or ⁇ -T7 antibodies.
  • D In vivo acetylation assay of endogenous p65. 293 cells were cotransfected with expression vectors encoding Tat (0.5 ⁇ g) and p300 (10 ⁇ g) as indicated and treated with TNF ⁇ (20 ng/ml) and trichostatin (TSA; 400 nM) over night.
  • TSA is an inhibitor of class I and II HDACs, but not sirtuins and was added to prolong nuclear retention of p65 through hyperacetylation of K218 and K220. Endogenous p65 was immunoprecipitated using ⁇ -p65 antibody and subjected to western blotting with ⁇ -AcK310 p65 or ⁇ -p65 antibodies.
  • Tat mutants deficient in the induction of p65 hyperacetylation were identified. These point mutants were expressed in 293 cells together with p65 and the p300 acetyltransferase. Expression of p300 induced acetylation of K310 in p65, and wild type Tat (two-exon, 101 aa) proteins further enhanced this acetylation ( FIG. 6A , lanes 1-3).
  • lentiviral vectors expressing wild type or mutant Tat proteins K41A and K50/51 A Tat. Because both Tat mutants have reduced transcriptional activities on the HIV LTR, we chose a lentiviral construct, which contained the heterologous elongation factor 1 ⁇ (EF-1 ⁇ ) promoter driving Tat and GFP expression instead of the HIV LTR. High titer viral stocks were produced and used to infect Jurkat T cells. Flow cytometry of GFP showed equivalent infection with wild type or mutant Tat-expressing viruses ( FIG. 6B ). Following treatment with ⁇ -CD3/28 antibodies, IL-2 mRNA levels were superinduced 2-3 fold by wild type Tat ( FIG. 6C ).
  • EF-1 ⁇ heterologous elongation factor 1 ⁇
  • FIGS. 6A-C Hyperacetylation of p65 by Tat contributes to T-cell hyperactivation (A) Induction of p65 hyperacetylation by wild type and mutant Tat.
  • Expression vectors encoding T7-tagged p65 (0.5 ⁇ g), p300 (2 ⁇ g), wild type and mutant Tat (1 ⁇ g each) were transiently cotransfected in 293 cells as indicated. Immunoprecipitations were performed with ⁇ -T7 agarose or ⁇ -FLAG agarose to isolate p65 or Tat followed by western blotting with ⁇ -AcK310 p65, ⁇ -T7, and ⁇ -FLAG antibodies.
  • (B) Flow cytometry of Jurkat T cells infected with lentiviral vectors expressing wild type or mutant Tat and GFP. Data are representative of three independent experiments in which infection efficiencies ranged from 75-85% GFP cells.
  • (C) Real-time RT-PCR analysis of IL-2 mRNA levels induced in infected Jurkat T cells after activation with ⁇ -CD3/28 antibodies. Data are presented as fold induction by ⁇ -CD3/28 treatment relative to vector (EF-1 ⁇ -GFP)-infected cells (100%). The average (mean ⁇ SEM) of three independent experiments is shown.
  • Tat binds to the SIRT1 catalytic site and impairs the ability of SIRT1 to deacetylate K310 in p65. It was also found that NF- ⁇ B activity is “superactivated” in the presence of Tat. Based on these results, a model is proposed where Tat hyperactivates T cells through inhibition of SIRT1 and abnormally sustained action of NF- ⁇ B. In the absence of Tat, a balance of p300 and SIRT1 activities regulates p65 acetylation. This balance tightly controls the activation of cellular genes involved in the immune response of T cells ( FIG. 7A ).
  • Tat expression disrupts the balance between p300 and SIRT1 and increases levels of acetylated p65 through inhibition of SIRT1 ( FIG. 7B ). This increase hyperactivates the function of NF- ⁇ B and the expression of NF- ⁇ B-responsive genes including IL-2.
  • NF- ⁇ B-responsive genes One of the most studied NF- ⁇ B-responsive genes is HIV itself.
  • the action of cellular transcription factors, most importantly NF- ⁇ B is essential for the production of full-length viral transcripts necessary for the initial synthesis of Tat.
  • Tat Once Tat accumulates to sufficient levels, it binds to TAR RNA and dramatically increases the elongation competence of the RNA polymerase II complex through recruitment of the P-TEFb complex. At that time, Tat may also enhance NF- ⁇ B function through inhibition of SIRT1 ( FIG. 7B ).
  • FIGS. 7A and 7B Tat blocks the SIRT1 deacetylase activity and superinduces T-cell activation and HIV transcription via NF- ⁇ B.
  • SIRT1 Activators Suppress Tat-Induced T Cell Hyperactivation
  • Lentiviral vectors which contained the heterologous elongation factor 1 ⁇ (EF-1 ⁇ ) promoter driving Tat and GFP expression, were generated. High titer viral stocks were produced and used to infect Jurkat T cells. Flow cytometry of GFP showed equivalent infection with EF-1 ⁇ -GFP or EF-1 ⁇ -Tat-GFP viruses. Infected cultures were stimulated with antibodies specific for the CD3 receptor and treated with increased concentrations of resveratrol (0.4 ⁇ M, 2 ⁇ M, and 10 ⁇ M), a known SIRT1 activator for 16 hrs. IL-2 protein was measured in the culture supernatant by ELISA assay.
  • IL-2 protein was superinduced ⁇ 4-fold by wild type Tat ( FIG. 8A ).
  • Tat In cells expressing Tat, ⁇ 2300 pg/ml of IL-2 protein was detected in response to ⁇ -CD3 treatment, whereas in control cells, only ⁇ 600 pg/ml of IL-2 protein was induced.
  • Superinduction of IL-2 in Tat-expressing cells was abolished by resveratrol in a concentration dependent manner ( FIG. 8A ). Treatment of infected cells with 10 ⁇ M of resveratrol completely impaired the ability of Tat to superinduce IL-2 production.
  • Resveratrol a polyphenolic compound present in red wine, functions as a (weak) reversible activator of SIRT1 enzymatic activity (Howitz et al., (2003) Nature 425:191-196., Baur et al., (2006) Nat. Rev. Drug. Discov. ). Recently, novel 1000-fold more potent and structurally unrelated small molecule activators of SIRT1 have been developed and have shown potent activity in obesity and insulin resistance (Milne et al., (2007) Nature 450:712-716).
  • FIGS. 8A and 8B Resveratrol and small molecule activator of SIRT1 (SRT2183) suppresses T cell hyperactivation by Tat.
  • SRT2183 small molecule activator of SIRT1
  • IL-2 protein levels measured by ELISA in the supernatant of infected cultures. 24 hrs after infection with EF1 ⁇ -GFP and EF1 ⁇ -Tat-GFP viruses, Jurkat T cells were stimulated with ⁇ -CD3 antibody in the presence of increased concentrations of resveratrol (0.4, 2, and 10 ⁇ M) for 16 hrs.
  • SIRT1 Regulates FoxP3 Stability and Treg Differentiation Through Deacetylation of Three Novel Acetylation Sites in FoxP3
  • the FoxP3 transcription factor is the master regulator of regulatory T cell (Treg) differentiation.
  • High-level FoxP3 expression is required to support the immune suppressive activity of Tregs while Tregs expressing low levels of FoxP3 may cause autoimmunity.
  • FoxP3 protein expression is regulated at the posttranscriptional level by the NAD+-dependent SIRT1 deacetylase. Induced Treg differentiation is suppressed when SIRT1 is overexpressed.
  • SIRT1 knockdown or treatment with a small molecule inhibitor of SIRT1 (Ex-527) stabilizes FoxP3 protein expression.
  • Treatment with Ex-527 prevents loss of FoxP3 expression in thymus-derived natural Tregs during ex vivo expansion.
  • SIRT1 Plays a Negative Role in the Differentiation of Induced Tregs (iTregs)
  • SIRT1 protein expression during ex vivo Treg differentiation was analyzed.
  • Na ⁇ ve CD4 + T cells (CD4+CD25 ⁇ CD69 ⁇ CD44 ⁇ ) were isolated from mouse spleen and lymph nodes and were activated by anti-CD3/CD28 antibodies alone or in combination with cytokines driving differentiation into T helper (Th) 1, Th2 or induced Treg (iTreg) cells.
  • Th T helper
  • iTreg induced Treg
  • SIRT1 plays a role in iTreg differentiation.
  • Activated CD4+ cells were infected with bicistronic retroviral vectors expressing SIRT1 and the Thy1.1 surface marker (MSCV-SIRT1-Thy1.1) or a control vector expressing Thy1.1 alone (MSCV-Thy1.1).
  • Cell were further differentiated into Tregs in the presence of recombinant human TGF- ⁇ and recombinant human IL-2 as previously described (Fu et al., 2004). After 3 days, cells were stained for surface Thy1.1 and intracellular FoxP3 and analyzed by flow cytometry.
  • nicotinamide-induced hyperacetylation of FoxP3 was more pronounced than hyperacetylation induced by trichostatin A (TSA), a class I and II HDAC inhibitor previously shown to increase FoxP3 acetylation (Li et al., 2007; Tao et al., 2007) ( FIG. 10C , lane 2).
  • TSA trichostatin A
  • acetylation of another transcription factor, AML1/Runx1 was increased to the same extent by TSA and nicotinamide treatment and both agents synergized to further increase acetylation ( FIG. 10D ).
  • SIRT1 Destabilizes the FoxP3 Protein Via K31, K262, and K267
  • Ex-527 was added to na ⁇ ve CD4+ T cells undergoing differentiation into iTreg cells.
  • the number of FoxP3-expressing cells increased up to 3-fold in the presence of Ex-527 as compared to cells treated with the drug vehicle alone ( FIG. 12A ).
  • MFI mean fluorescent intensity
  • GFP+ cells were isolated from GFP-Foxp3 knock-in mice, in which green fluorescent protein (GFP) is expressed as a fusion protein with FoxP3.
  • GFP green fluorescent protein
  • Total CD4+ T cells were isolated from spleen and lymph nodes of these mice, and GFP+ T cells were sorted with >95% purity.
  • GFP+ T cells were expanded by anti-CD3/CD28 beads and IL-2 for 5 days when Ex-527 or dimethylsulfoxide (DMSO) was added. Two days later, 75% of cells were GFP-positive regardless whether Ex-527 was added or not; however, the MFI of GFP was increased in Ex-527-treated cells ( FIG. 12B , day 7).
  • SIRT1 inhibitors e.g., Ex-527
  • FIGS. 9A and 9B SIRT1 Plays a Negative Role in iTreg Differentiation
  • FIG. 1 A Western blot analysis of SIRT1 in mouse CD4+ T cells. Na ⁇ ve CD4+ T cells isolated from mouse spleen and lymph node were activated with CD3 and CD28 antibodies and differentiated into Th1, Th2 and iTreg cells as described in material and methods.
  • FIG. 1 A Western blot analysis of SIRT1 in mouse CD4+ T cells. Na ⁇ ve CD4+ T cells isolated from mouse spleen and lymph node were activated with CD3 and CD28 antibodies and differentiated into Th1, Th2 and iTreg cells as described in material and methods.
  • FIG. 1 A and B show one representative experiment of two with the same results.
  • FIGS. 10A-D SIRT1 Interacts with FoxP3 and Inhibition of SIRT1 Deacetylase Activity Increases Acetylation of FoxP3.
  • (A) Communoprecipitation/western blot analysis of FLAG-tagged FoxP3 and HA-tagged SIRT1 after transfection of corresponding expression vectors or empty vector controls into 293T cells.
  • (B) Communoprecipitation assay of SIRT1 and FoxP3 deletion mutants. A schematic summary of the binding of FoxP3 mutants to SIRT1 is shown. The forkhead box (FH) indicates the DNA binding domain ZnF, zinc finger domain; LZ, leucine zipper.
  • (C) Immunoprecipitation/Western blot analysis of acetylated FLAG-FoxP3 in response to trichostatin A (TSA) or nicotinamide in 293 T cells.
  • TSA trichostatin A
  • FIGS. 11A-D SIRT1 destabilizes FoxP3 protein through deacetylation of three newly identified acetylation sites
  • A Western blot analysis of FLAG-FoxP3 or FLAG-AML1/Runx1in 293 T cells treated with nicotinamide (5 mM), Ex-527 (50 ⁇ M) or trichostatin A TSA (400 nM) for 16 hours.
  • B Scheme of acetylated lysine residues identified by mass spectrometry using immunoprecipitated FLAG-FoxP3 protein. The murine protein is shown.
  • C Western blot analysis of wild type or mutant FLAG-FoxP3 proteins in 293 T cells treated with Ex-527 (50 ⁇ M) or DMSO as a control.
  • D Western blot analysis of wild type or mutant K31/K262/K267R (3KR) FLAG-FoxP3 in 293 cells treated with cycloheximide (CHX) and/or Ex-527 for indicated times.
  • FIGS. 12A and 12B Inhibition of SIRT1 activity by Ex-527 promotes differentiation of iTreg cells and FoxP3 expression in nTreg cells.
  • GFP+nTregs were expanded by ⁇ -CD3/CD28 beads and hIL-2 (2000 U/ml) for 5 days. Cells were further cultured with either DMSO or Ex-527 for additional 2 and 7 days and FoxP3 expression was analyzed as in (A). Western blot analysis of FoxP3 in cell lysates from GFP ⁇ , GFP+, and GFP+ cultures expanded as described in the presence of DMSO or EX-527.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Transplantation (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an inhibitor of SIRT1. The present disclosure provides a method of reducing an immune response, e.g., to treat chronic immune hyperactivity, the method generally involving administering to an individual in need thereof an activator of SIRT1. The present disclosure provides a method of modulating activation and differentiation of CD4+ T cells.

Description

    CROSS-REFERENCE
  • This application claims the benefit of U.S. Provisional Patent Application No. 61/026,997, filed Feb. 7, 2008, and is a continuation-in part of International Patent Application No. PCT/US2009/000761, which applications are incorporated herein by reference in their entirety.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
  • This invention was made with government support under grant no. R01 A1067118-01A awarded by the National Institutes of Health. The government has certain rights in the invention.
  • BACKGROUND
  • Immune activation is a hallmark of human immunodeficiency virus-1 (HIV-1) infection and a significant factor that promotes continuous viral replication and CD4+ T-cell depletion. In HIV-infected individuals, levels of circulating activation markers correlate with accelerated disease progression and shortened survival. HIV infection is critically dependent on the activated state of CD4+ T cells since the virus cannot replicate efficiently in resting T cells. Quiescent T cells in blood are refractory to infection because of blocks at the level of reverse transcription and proviral integration. In addition, T-cell activation enhances viral transcription through the activation of various transcription factors, notably nuclear factor κB (NF-κB).
  • SIRT1 is a mammalian homologue of the yeast transcriptional repressor silent information regulator 2 (Sir2), an important factor governing longevity in yeast. Like Sir2, SIRT1 requires nicotinamide adenine dinucleotide (NAD+) as a cofactor, which links its activity to the metabolic state of the cell. In addition to its enzymatic activity on histone substrates in vitro, recent experimental evidence suggests that SIRT1 predominantly targets nonhistone proteins for deacetylation. It has been reported that Tat is a substrate for the deacetylase activity of SIRT1. Acetylation of lysine 50 (K50) in Tat is mediated by the histone acetyltransferase activities of p300 and human GCNS and generates binding sites for the bromodomains of PCAF and Brg1. SIRT1 binds and deacetylates Tat at K50, a process necessary to recycle nonacetylated Tat protein for binding to TAR RNA and the cellular positive transcription elongation factor b (P-TEFb).
  • LITERATURE
    • Milne et al., (2007) Nature 450:712-716; U.S. Patent Publication No. 2007/0043050; U.S. Pat. No. 7,345,178; Ott et al. (1997) Science 275:1481; U.S. Patent Publication No. 2009/0012080; U.S. Patent Publication No. 2008/0255382; Nayagam et al. (2006) J. Biomol. Screening 11:959; U.S. Patent Publication No. 2007/0105109; U.S. Patent Publication No. 2007/0190073; U.S. Patent Publication No. 2005/0209300.
    SUMMARY OF THE INVENTION
  • The present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an inhibitor of SIRT1. The present disclosure provides a method of reducing an immune response, e.g., to treat chronic immune hyperactivity, the method generally involving administering to an individual in need thereof an activator of SIRT1. The present disclosure provides a method of modulating activation and differentiation of CD4+ T cells.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-F depict data showing the effect of Tat and nicotinamide on T cell hyperactivation.
  • FIGS. 2A-D depict the Tat-mediated superinduction of NF-κB-responsive genes.
  • FIGS. 3A-C depicts the effect of Tat on the inhibitory effect of SIRT1 on NK-κ-B-responsive promoters.
  • FIGS. 4A-E depict binding of Tat to the acetyl lysine-binding site in SIRT1.
  • FIGS. 5A-D depict the effect of Tat on SIRT1 deacetylase activity.
  • FIGS. 6A-C depict the effect of hyperacetylation of p65 by Tat on T-cell hyperactivation.
  • FIGS. 7A and 7B depict a model for Tat effects on SIRT1 deacetylase activity, T-cell activation, and HIV transcription.
  • FIGS. 8A and 8B depict the effect of SIRT1 activators on Tat-mediated T cell hyperactivation.
  • FIGS. 9A and 9B depict the effect of SIRT1 on induced Treg (iTreg) differentiation.
  • FIGS. 10A-D depict interaction of SIRT1 with FoxP3, and the effect of inhibition of SIRT1 deacetylase activity on acetylation of FoxP3.
  • FIGS. 11A-D depict destabilization of FoxP3 protein via deacetylation by SIRT1.
  • FIGS. 12A and 12B depict the effect of inhibition of SIRT1 activity on differentiation of iTreg cells and FoxP3 expression in nTreg cells.
  • FIG. 13 depicts an alignment of amino acid sequences of human FoxP3 (GenBank Accession Nos. NP054728 and NM014009), mouse FoxP3 (GenBank Accession Nos. NM054039 and NP473380), and cow FoxP3 (GenBank Accession Nos. NM001045933 and NP001039398).
  • DEFINITIONS
  • As used herein, the terms “treatment,” “treating,” and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease. “Treatment”, as used herein, covers any treatment of a disease in a mammal, e.g., in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, e.g., causing regression of the disease, e.g., to completely or partially remove symptoms of the disease.
  • The term “effective amount” or “therapeutically effective amount” means a dosage sufficient to provide for treatment for the disease state being treated or to otherwise provide the desired effect (e.g., induction of an effective immune response, reduction of chronic immune hyperactivity, etc.). The precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., weight, age, etc.), the disease, and the treatment being effected.
  • The terms “individual,” “host,” “subject,” and “patient,” used interchangeably herein, refer to a mammal, including, but not limited to, murines, felines, simians, humans, etc. In some embodiments, an individual is a human. In some embodiments, an individual is a rodent (e.g., a mouse, a rat, etc.) or a lagomorph.
  • A “pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” and “pharmaceutically acceptable adjuvant” means an excipient, diluent, carrier, or adjuvant that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, or adjuvant that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable excipient, diluent, carrier and adjuvant” as used in the specification and claims includes one and more than one such excipient, diluent, carrier, and adjuvant.
  • As used herein, a “pharmaceutical composition” is meant to encompass a composition suitable for administration to a subject, such as a mammal, e.g., a human. In general a “pharmaceutical composition” is sterile, and is free of contaminants that are capable of eliciting an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade). Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, intratracheal and the like. In some embodiments the composition is suitable for administration by a transdermal route, using a penetration enhancer other than dimethylsulfoxide (DMSO). In other embodiments, the pharmaceutical compositions are suitable for administration by a route other than transdermal administration. A pharmaceutical composition will in some embodiments include a compound and a pharmaceutically acceptable excipient. In some embodiments, a pharmaceutically acceptable excipient is other than DMSO.
  • As used herein, “pharmaceutically acceptable derivatives” of a compound of the invention include salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives may be readily prepared by those of skill in this art using known methods for such derivatization. The compounds produced may be administered to animals or humans without substantial toxic effects and are either pharmaceutically active or are prodrugs.
  • A “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4′-methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
  • Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
  • Unless defined otherwise, 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 invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
  • It must be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a SIRT1 modulator” includes a plurality of such modulators and reference to “the SIRT1 inhibitor” includes reference to one or more SIRT1 inhibitors and equivalents thereof known to those skilled in the art, and so forth. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation.
  • The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.
  • DETAILED DESCRIPTION
  • The present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an inhibitor of SIRT1. The present disclosure provides a method of reducing an immune response (e.g., to treat chronic immune hyperactivity, to treat an autoimmune disorder), the method generally involving administering to an individual in need thereof an activator of SIRT1. The present disclosure provides a method of modulating activation and differentiation of CD4+ T cells, the method generally involving contacting the CD4+ T cell with a SIRT1 inhibitor.
  • As used herein, “SIRT1” refers to a polypeptide exhibiting nicotinamide adenosine dinucleotide (NAD)-dependent deacetylase activity, and having an amino acid sequence that is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, identical to the amino acid sequence of a known SIRT1 polypeptide. A SIRT1 protein includes yeast Sir2 (GenBank Accession No. P53685); C. elegans Sir-2.1 (GenBank Accession No. NP501912); human SIRT1 (see, e.g., GenBank Accession No. NM012238 or NP036370 (or AF083106); Frye ((1999) Biochem. Biophys. Res. Comm. 260:273); and GenBank Accession Nos. Q96EB6, AAH12499, NP036370, and AAD40849); mouse SIR1 (see, e.g., GenBank Accession Nos. Q923E4 and NP062786); and equivalents and fragments thereof. For example, a SIRT1 polypeptide comprises an amino acid sequence having at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, amino acid sequence identity with a contiguous stretch of from about 500 amino acids (aa) to about 550 aa, from about 550 aa to about 600 aa, from about 600 aa to about 650 aa, from about 650 aa to about 700 aa, or from about 700 aa to about 747 aa, of the amino acid sequence set forth in SEQ ID NO:1.
  • SIRT1 Modulators
  • A suitable SIRT1 modulator modulates an enzymatic activity of a SIRT1 polypeptide.
  • For example, a suitable SIRT1 modulator modulates Tat deacetylase activity of SIRT1. SIRT1 Tat deacetylase activity can be determined using known methods. For example, a Tat polypeptide (e.g, a synthetic Tat polypeptide) that is acetylated is used as a substrate. A reaction sample can include an acetylated Tat polypeptide, NAD+, a SIRT1 polypeptide, and a SIRT1 modulator. Acetylated Tat polypeptides include Tat polypeptides having one or more acetylated lysine residues, e.g., acetylated K28, acetylated K41, and acetylated K50. A suitable acetylated Tat polypeptide includes the amino acid sequence Ser-Tyr-Gly-Arg-AcLys-Lys-Lys-Arg-Arg-Gln-Arg (SEQ ID NO:02), where AcLys is acetylated lysine. An acetylated Tat protein can be generated as described in, e.g., Doerr et al. (2002) EMBO J. 21:2715-2723; or Peloponese (1999) J. Biol. Chem. 274:11473-11478. Suitable acetylated Tat polypeptides are described in, e.g., U.S. Pat. No. 7,485,416 and U.S. Patent Publication No. 2005/0287597.
  • The effect of the SIRT1 modulator on SIRT1 Tat deacetylase activity can be determined by measuring the amount of deacetylated Tat polypeptide produced by action of the SIRT1 polypeptide in the presence of the SIRT1 modulator, compared to the amount of deacetylated Tat polypeptide produced by action of the SIRT1 polypeptide in a control reaction sample that does not include the SIRT1 modulator. As an alternative to, or in addition to, measuring the amount of deacetylated Tat polypeptide, the amount of remaining acetylated Tat can be measured. Methods of determining the level of acetylated Tat in a sample include immunological assays using antibody that is specific for acetylated form of Tat, and that therefore distinguishes between acetylated Tat and deacetylated Tat. Any of a variety of immunological assays can be used, including, e.g., enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), protein blot (“Western” blot) assays, and the like. In some embodiments, mass spectroscopy is used. SIRT1 activity can also be determined by measuring the level of NAD in the test sample. The action of SIRT1 on acetylated Tat can be coupled to a second enzymatic reaction that reduces NAD to NADH, and measuring fluorescence of NADH at, e.g., 340 nm. See U.S. Pat. No. 7,485,416 and U.S. Patent Publication No. 2005/0287597 for descriptions of methods of determining SIRT1 Tat deacetylase activity.
  • In some embodiments, a suitable SIRT1 modulator (a SIRT1 activator or a SIRT1 inhibitor) is a selective SIRT1 modulator. For example, in some embodiments, a suitable SIRT1 activator is a selective SIRT1 activator; and in some embodiments, a suitable SIRT inhibitor is a selective SIRT1 inhibitor. For example, in some embodiments, a suitable SIRT1 activator increases the enzymatic activity of a SIRT1 polypeptide, but does not substantially increase the enzymatic activity of any other sirtuin. For example, in some embodiments, a suitable SIRT1 activator increases the enzymatic activity of a SIRT1 polypeptide, but does not substantially increase the enzymatic activity of SIRT2, SIRT4, or SIRT5. In some embodiments, a suitable SIRT1 inhibitor reduces the enzymatic activity of a SIRT1 polypeptide but does not substantially reduce the enzymatic activity of any other sirtuin. For example, in some embodiments, a suitable SIRT1 inhibitor reduces the enzymatic activity of a SIRT1 polypeptide, but does not substantially reduce the enzymatic activity of SIRT2, SIRT4, or SIRT5.
  • In some embodiments, a suitable SIRT1 modulator modulates the activity of SIRT1, and can also modulate the activity of one or more additional sirtuins. For example, in some embodiments, a suitable SIRT1 activator increases the enzymatic activity of a SIRT1 polypeptide, and can also increase the enzymatic activity of SIRT5. As another example, in some embodiments, a suitable SIRT1 inhibitor reduces the enzymatic activity of a SIRT1 polypeptide, and can also reduce the enzymatic activity of SIRT5.
  • A suitable SIRT1 activator can increase the enzymatic activity of SIRT1 by at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 20-fold, or more than 20-fold. A suitable SIRT1 activator can increase the enzymatic activity of SIRT1 by from about 25% to about 50%, from about 50% to about 75%, from about 75% to about 2-fold, from about 2-fold to about 5-fold, from about 5-fold to about 10-fold, from about 10-fold to about 20-fold, or more than 20-fold.
  • A suitable SIRT1 inhibitor can reduce the enzymatic activity of SIRT1 by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or more than 75%. A suitable SIRT1 inhibitor can reduce the enzymatic activity of SIRT1 by from about 5% to about 10%, from about 10% to about 15%, from about 15% to about 20%, from about 20% to about 25%, from about 25% to about 30%, from about 35% to about 40%, from about 40% to about 50%, from about 50% to about 60%, from about 60% to about 75%, or more than 75%.
  • A suitable SIRT1 activator can increase SIRT1 enzymatic activity at an EC50 (half maximal effective concentration) of from about 1 nM to about 1 mM, e.g., from about 1 nM to about 10 nM, from about 10 nM to about 15 nM, from about 15 nM to about 25 nM, from about 25 nM to about 50 nM, from about 50 nM to about 75 nM, from about 75 nM to about 100 nM, from about 100 nM to about 150 nM, from about 150 nM to about 200 nM, from about 200 nM to about 250 nM, from about 250 nM to about 300 nM, from about 300 nM to about 350 nM, from about 350 nM to about 400 nM, from about 400 nM to about 450 nM, from about 450 nM to about 500 nM, from about 500 nM to about 750 nM, from about 750 nM to about 1 μM, from about 1 μM to about 10 μM, from about 10 μM to about 25 μM, from about 25 μM to about 50 μM, from about 50 μM to about 75 μM, from about 75 μM to about 100 μM, from about 100 μM to about 250 μM, from about 250 μM to about 500 μM, or from about 500 μM to about 1 mM.
  • A suitable SIRT1 activator can increase SIRT1 enzymatic activity at an EC1.5 (concentration of compound required to increase enzyme activity by 50%) of from about 1 nM to about 1 mM, e.g., from about 1 nM to about 10 nM, from about 10 nM to about 15 nM, from about 15 nM to about 25 nM, from about 25 nM to about 50 nM, from about 50 nM to about 75 nM, from about 75 nM to about 100 nM, from about 100 nM to about 150 nM, from about 150 nM to about 200 nM, from about 200 nM to about 250 nM, from about 250 nM to about 300 nM, from about 300 nM to about 350 nM, from about 350 nM to about 400 nM, from about 400 nM to about 450 nM, from about 450 nM to about 500 nM, from about 500 nM to about 750 nM, from about 750 nM to about 1 μM, from about 1 μM to about 10 μM, from about 10 μM to about 25 μM, from about 25 μM to about 50 μM, from about 50 μM to about 75 μM, from about 75 μM to about 100 μM, from about 100 μM to about 250 μM, from about 250 μM to about 500 μM, or from about 500 μM to about 1 mM. For example, a suitable SIRT1 activator can have an EC1.5 of from about 0.01 μM to about 100 μM, e.g., from about 0.01 μM to about 0.1 μM, from about 0.1 μM to about 0.5 μM, from about 0.5 μM to about 1.0 μM, from about 1.0 μM to about 10 μM, from about 10 μM to about 25 μM, from about 25 μM to about 50 μM, or from about 50 μM to about 100 μM.
  • A suitable SIRT1 inhibitor can have an IC50 of less than 50 μM, e.g., a suitable SIRT1 inhibitor can have an IC50 of from about 50 μM to about 5 nm, or less than 5 nM. For example, in some embodiments, a suitable SIRT1 inhibitor has an IC50 of from about 50 μM to about 25 μM, from about 25 μM to about 10 μM, from about 10 μM to about 5 μM, from about 5 μM to about 1 μM, from about 1 μM to about 500 nM, from about 500 nM to about 400 nM, from about 400 nM to about 300 nM, from about 300 nM to about 250 nM, from about 250 nM to about 200 nM, from about 200 nM to about 150 nM, from about 150 nM to about 100 nM, from about 100 nM to about 50 nM, from about 50 nM to about 30 nM, from about 30 nM to about 25 nM, from about 25 nM to about 20 nM, from about 20 nM to about 15 nM, from about 15 nM to about 10 nM, from about 10 nM to about 5 nM, or less than about 5 nM.
  • SIRT1 modulators are known in the art, and any modulator of SIRT1 can be used. For example, Ota et al. ((2006) Oncogene 25:176) discusses sirtinol; resveratrol is discussed in, e.g., de la Lastra (2005) Mol. Nutr. Food Res. 49:405; Napper et al. ((2005) J. Med. Chem. 48:8045) discusses indole compounds that are SIRT1 inhibitors; Solomon et al. ((2006) Mol. Cell. Biol. 26:28) discusses EX-527, a SIRT1 inhibitor; U.S. Patent Publication Nos. 2007/0043050, 2007/0037865, and 2007/0037809 discuss SIRT1 modulators; and U.S. Patent Publication No. 2008/0021063 discusses SIRT1 modulators.
  • Whether a given agent (e.g., a SIRT1 modulator, such as a SIRT1 inhibitor or a SIRT1 activator) is effective in modulating an immune response (e.g., increasing an immune response; e.g., reducing an immune response, such as reducing chronic immune hyperactivity) can be readily determined using well known methods to assess one or more immune response parameters Immune response parameters include, but are not limited to, CD4 count; serum cytokine levels, e.g., serum levels of IL-2; serum antibody levels; levels of an autoantibody; and the like.
  • Methods of Reducing an Immune Response
  • In some embodiments, the present disclosure provides methods of reducing an immune response, the methods generally involving administering to an individual in need thereof an effective amount of a SIRT1 activator.
  • In some embodiments, an effective amount of a SIRT1 activator is an amount that, when administered to an individual in one or more doses, is effective to reduce the level of a cytokine associated with an immune response. Cytokines associated with an immune response include, e.g., interleukin-2 (IL-2), tumor necrosis factor-alpha (TNF-α), interleukin-4 (IL-4), interferon gamma (IFN-γ), and interleukin-12 (IL-12). In some embodiments, an effective amount of a SIRT1 activator is an amount that, when administered to an individual in one or more doses, is effective to reduce the level in the individual of a cytokine associated with an immune response by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%, compared to the level of the cytokine in the absence of treatment with the SIRT1 activator.
  • In some embodiments, an effective amount of a SIRT1 activator is an amount that, when administered to an individual in one or more doses, is effective to reduce the level of IL-2 (e.g., circulating IL-2, e.g., a serum level of IL-2) in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%, compared to the level of IL-2 in the individual in the absence of treatment with the SIRT1 activator.
  • Methods of reducing an immune response are useful for treating chronic immune hyperactivation. Thus, the instant disclosure provides methods for treating chronic immune hyperactivation, the methods generally involving administering to an individual in need thereof an effective amount of a SIRT1 activator. In some embodiments, the instant disclosure provides a method for treating chronic immune hyperactivation, the method comprising administering to an individual in need thereof an effective amount of a selective SIRT1 activator. In some embodiments, the instant disclosure provides a method for treating chronic immune hyperactivation that results from infection with an immunodeficiency virus, e.g., a human immunodeficiency virus (e.g., HIV-1), the method comprising administering to an individual in need thereof an effective amount of a SIRT1 activator, where in some embodiments, the SIRT1 inhibitor is a selective SIRT1 activator.
  • SIRT1 Activators
  • Examples of SIRT1 activators that are suitable for use in a subject method include, but are not limited to, resveratrol ((E)-5-(p-Hydroxystyryl)resorcinol (E)-5-(4-hydroxystyryl)benzene-1,3-diol); or 3,5,4′-trihydroxy-trans-stilbene); butein (3,4,2′,4′-tetrahydroxychalcone); piceatannol (3,5,3′,4′-tetrahydroxy-trans-stilbene); isoliquiritigenin (4,2′,4′-trihydroxychalcone); fisetin (3,7,3′,4′-tetrahydroxyflavone); quercetin (3,5,7,3′,4′-pentahydroxyflavone); a SIRT1 activator as described in U.S. Pat. No. 7,345,178; a SIRT1 activator as described in U.S. Patent Publication No. 2008/02555382; and a SIRT1 activator as described in U.S. Patent Publication No. 2009/0012080. Pharmaceutically acceptable salts of any of the foregoing SIRT1 activators are also suitable for use in a subject method.
  • For example, a suitable SIRT1 activator is a compound of any one of Formulas I-XXVIII as described in U.S. Pat. No. 7,345,178, where substituents are as described in U.S. Pat. No. 7,345,178, or a pharmaceutically acceptable salt of a compound of any one of Formulas I-XXVIII as described in U.S. Pat. No. 7,345,178, provided that the compound activates SIRT1 activity. For example, a suitable SIRT1 activator includes a compound shown in Table 4 of U.S. Pat. No. 7,345,178.
  • For example, suitable SIRT1 activators are shown in Table 1, below.
  • TABLE 1
    Figure US20140030295A1-20140130-C00001
    Compound 1
    Figure US20140030295A1-20140130-C00002
    Compound 2
    Figure US20140030295A1-20140130-C00003
    Compound 3
    Figure US20140030295A1-20140130-C00004
    Compound 4
    Figure US20140030295A1-20140130-C00005
    Compound 5
    Figure US20140030295A1-20140130-C00006
    Compound 6
    Figure US20140030295A1-20140130-C00007
    Compound 7
    Figure US20140030295A1-20140130-C00008
    Compound 8
    Figure US20140030295A1-20140130-C00009
    Compound 9
    Figure US20140030295A1-20140130-C00010
    Compound 10
    Figure US20140030295A1-20140130-C00011
    Compound 19
    Figure US20140030295A1-20140130-C00012
    Compound 20
    Figure US20140030295A1-20140130-C00013
    Compound 21
    Figure US20140030295A1-20140130-C00014
    Compound 22
    Figure US20140030295A1-20140130-C00015
    Compound 24
    Figure US20140030295A1-20140130-C00016
    Compound 27
    Figure US20140030295A1-20140130-C00017
    Compound 29
    Figure US20140030295A1-20140130-C00018
    Compound 31
    Figure US20140030295A1-20140130-C00019
    Compound 32
    Figure US20140030295A1-20140130-C00020
    Compound 33
    Figure US20140030295A1-20140130-C00021
    Compound 34
    Figure US20140030295A1-20140130-C00022
    Compound 35
    Figure US20140030295A1-20140130-C00023
    Compound 36
    Figure US20140030295A1-20140130-C00024
    Compound 37
    Figure US20140030295A1-20140130-C00025
    Compound 38
    Figure US20140030295A1-20140130-C00026
    Compound 39
    Figure US20140030295A1-20140130-C00027
    Compound 40
    Figure US20140030295A1-20140130-C00028
    Compound 41
    Figure US20140030295A1-20140130-C00029
    Compound 42
    Figure US20140030295A1-20140130-C00030
    Compound 43
    Figure US20140030295A1-20140130-C00031
    Compound 45
    Figure US20140030295A1-20140130-C00032
    Compound 46
    Figure US20140030295A1-20140130-C00033
    Compound 48
    Figure US20140030295A1-20140130-C00034
    Compound 49
    Figure US20140030295A1-20140130-C00035
    Compound 50
    Figure US20140030295A1-20140130-C00036
    Compound 51
    Figure US20140030295A1-20140130-C00037
    Compound 52
    Figure US20140030295A1-20140130-C00038
    Compound 53
    Figure US20140030295A1-20140130-C00039
    Compound 54
    Figure US20140030295A1-20140130-C00040
    Compound 55
    Figure US20140030295A1-20140130-C00041
    Compound 56
    Figure US20140030295A1-20140130-C00042
    Compound 57
    Figure US20140030295A1-20140130-C00043
    Compound 58
    Figure US20140030295A1-20140130-C00044
    Compound 59
    Figure US20140030295A1-20140130-C00045
    Compound 60
    Figure US20140030295A1-20140130-C00046
    Compound 61
    Figure US20140030295A1-20140130-C00047
    Compound 62
    Figure US20140030295A1-20140130-C00048
    Compound 63
    Figure US20140030295A1-20140130-C00049
    Compound 64
    Figure US20140030295A1-20140130-C00050
    Compound 65
    Figure US20140030295A1-20140130-C00051
    Compound 66
    Figure US20140030295A1-20140130-C00052
    Compound 67
    Figure US20140030295A1-20140130-C00053
    Compound 68
    Figure US20140030295A1-20140130-C00054
    Compound 69
    Figure US20140030295A1-20140130-C00055
    Compound 70
    Figure US20140030295A1-20140130-C00056
    Compound 71
    Figure US20140030295A1-20140130-C00057
    Compound 72
    Figure US20140030295A1-20140130-C00058
    Compound 73
    Figure US20140030295A1-20140130-C00059
    Compound 74
    Figure US20140030295A1-20140130-C00060
    Compound 75
    Figure US20140030295A1-20140130-C00061
    Compound 76
    Figure US20140030295A1-20140130-C00062
    Compound 77
    Figure US20140030295A1-20140130-C00063
    Compound 78
    Figure US20140030295A1-20140130-C00064
    Compound 79
    Figure US20140030295A1-20140130-C00065
    Compound 80
    Figure US20140030295A1-20140130-C00066
    Compound 81
    Figure US20140030295A1-20140130-C00067
    Compound 82
    Figure US20140030295A1-20140130-C00068
    Compound 83
    Figure US20140030295A1-20140130-C00069
    Compound 84
    Figure US20140030295A1-20140130-C00070
    Compound 85
    Figure US20140030295A1-20140130-C00071
    Compound 86
    Figure US20140030295A1-20140130-C00072
    Compound 87
    Figure US20140030295A1-20140130-C00073
    Compound 88
    Figure US20140030295A1-20140130-C00074
    Compound 89
    Figure US20140030295A1-20140130-C00075
    Compound 90
    Figure US20140030295A1-20140130-C00076
    Compound 91
    Figure US20140030295A1-20140130-C00077
    Compound 92
    Figure US20140030295A1-20140130-C00078
    Compound 93
    Figure US20140030295A1-20140130-C00079
    Compound 94
    Figure US20140030295A1-20140130-C00080
    Compound 95
    Figure US20140030295A1-20140130-C00081
    Compound 96
    Figure US20140030295A1-20140130-C00082
    Compound 97
    Figure US20140030295A1-20140130-C00083
    Compound 98
    Figure US20140030295A1-20140130-C00084
    Compound 99
    Figure US20140030295A1-20140130-C00085
    Compound 100
    Figure US20140030295A1-20140130-C00086
    Compound 101
    Figure US20140030295A1-20140130-C00087
    Compound 102
    Figure US20140030295A1-20140130-C00088
    Compound 103
    Figure US20140030295A1-20140130-C00089
    Compound 104
    Figure US20140030295A1-20140130-C00090
    Compound 105
    Figure US20140030295A1-20140130-C00091
    Compound 106
    Figure US20140030295A1-20140130-C00092
    Compound 107
    Figure US20140030295A1-20140130-C00093
    Compound 108
    Figure US20140030295A1-20140130-C00094
    Compound 109
    Figure US20140030295A1-20140130-C00095
    Compound 110
    Figure US20140030295A1-20140130-C00096
    Compound 111
    Figure US20140030295A1-20140130-C00097
    Compound 112
    Figure US20140030295A1-20140130-C00098
    Compound 113
    Figure US20140030295A1-20140130-C00099
    Compound 114
    Figure US20140030295A1-20140130-C00100
    Compound 115
    Figure US20140030295A1-20140130-C00101
    Compound 116
    Figure US20140030295A1-20140130-C00102
    Compound 117
    Figure US20140030295A1-20140130-C00103
    Compound 118
    Figure US20140030295A1-20140130-C00104
    Compound 119
    Figure US20140030295A1-20140130-C00105
    Compound 120
    Figure US20140030295A1-20140130-C00106
    Compound 121
    Figure US20140030295A1-20140130-C00107
    Compound 122
    Figure US20140030295A1-20140130-C00108
    Compound 123
    Figure US20140030295A1-20140130-C00109
    Compound 124
    Figure US20140030295A1-20140130-C00110
    Compound 125
    Figure US20140030295A1-20140130-C00111
    Compound 126
    Figure US20140030295A1-20140130-C00112
    Compound 127
    Figure US20140030295A1-20140130-C00113
    Compound 128
    Figure US20140030295A1-20140130-C00114
    Compound 129
    Figure US20140030295A1-20140130-C00115
    Compound 130
    Figure US20140030295A1-20140130-C00116
    Compound 131
    Figure US20140030295A1-20140130-C00117
    Compound 132
    Figure US20140030295A1-20140130-C00118
    Compound 133
    Figure US20140030295A1-20140130-C00119
    Compound 134
    Figure US20140030295A1-20140130-C00120
    Compound 135
    Figure US20140030295A1-20140130-C00121
    Compound 136
    Figure US20140030295A1-20140130-C00122
    Compound 137
    Figure US20140030295A1-20140130-C00123
    Compound 138
    Figure US20140030295A1-20140130-C00124
    Compound 139
    Figure US20140030295A1-20140130-C00125
    Compound 141
    Figure US20140030295A1-20140130-C00126
    Compound 142
    Figure US20140030295A1-20140130-C00127
    Compound 143
    Figure US20140030295A1-20140130-C00128
    Compound 144
    Figure US20140030295A1-20140130-C00129
    Compound 145
    Figure US20140030295A1-20140130-C00130
    Compound 146
    Figure US20140030295A1-20140130-C00131
    Compound 147
    Figure US20140030295A1-20140130-C00132
    Compound 148
    Figure US20140030295A1-20140130-C00133
    Compound 149
    Figure US20140030295A1-20140130-C00134
    Compound 150
    Figure US20140030295A1-20140130-C00135
    Compound 151
    Figure US20140030295A1-20140130-C00136
    Compound 152
    Figure US20140030295A1-20140130-C00137
    Compound 153
    Figure US20140030295A1-20140130-C00138
    Compound 154
    Figure US20140030295A1-20140130-C00139
    Compound 155
    Figure US20140030295A1-20140130-C00140
    Compound 156
    Figure US20140030295A1-20140130-C00141
    Compound 157
    Figure US20140030295A1-20140130-C00142
    Compound 158
    Figure US20140030295A1-20140130-C00143
    Compound 159
    Figure US20140030295A1-20140130-C00144
    Compound 160
    Figure US20140030295A1-20140130-C00145
    Compound 161
    Figure US20140030295A1-20140130-C00146
    Compound 162
    Figure US20140030295A1-20140130-C00147
    Compound 163
    Figure US20140030295A1-20140130-C00148
    Compound 164
    Figure US20140030295A1-20140130-C00149
    Compound 165
    Figure US20140030295A1-20140130-C00150
    Compound 166
    Figure US20140030295A1-20140130-C00151
    Compound 167
    Figure US20140030295A1-20140130-C00152
    Compound 168
    Figure US20140030295A1-20140130-C00153
    Compound 169
    Figure US20140030295A1-20140130-C00154
    Compound 174
    Figure US20140030295A1-20140130-C00155
    Compound 175
    Figure US20140030295A1-20140130-C00156
    Compound 176
    Figure US20140030295A1-20140130-C00157
    Compound 177
    Figure US20140030295A1-20140130-C00158
    Compound 178
    Figure US20140030295A1-20140130-C00159
    Compound 179
    Figure US20140030295A1-20140130-C00160
    Compound 180
    Figure US20140030295A1-20140130-C00161
    Compound 181
    Figure US20140030295A1-20140130-C00162
    Compound 182
    Figure US20140030295A1-20140130-C00163
    Compound 183
    Figure US20140030295A1-20140130-C00164
    Compound 184
    Figure US20140030295A1-20140130-C00165
    Compound 185
    Figure US20140030295A1-20140130-C00166
    Compound 186
    Figure US20140030295A1-20140130-C00167
    Compound 187
    Figure US20140030295A1-20140130-C00168
    Compound 188
    Figure US20140030295A1-20140130-C00169
    Compound 189
    Figure US20140030295A1-20140130-C00170
    Compound 190
    Figure US20140030295A1-20140130-C00171
    Compound 191
    Figure US20140030295A1-20140130-C00172
    Compound 192
    Figure US20140030295A1-20140130-C00173
    Compound 193
    Figure US20140030295A1-20140130-C00174
    Compound 194
    Figure US20140030295A1-20140130-C00175
    Compound 195
    Figure US20140030295A1-20140130-C00176
    Compound 196
    Figure US20140030295A1-20140130-C00177
    Compound 197
    Figure US20140030295A1-20140130-C00178
    Compound 198
    Figure US20140030295A1-20140130-C00179
    Compound 199
    Figure US20140030295A1-20140130-C00180
    Compound 200
    Figure US20140030295A1-20140130-C00181
    Compound 201
    Figure US20140030295A1-20140130-C00182
    Compound 202
    Figure US20140030295A1-20140130-C00183
    Compound 203
    Figure US20140030295A1-20140130-C00184
    Compound 204
    Figure US20140030295A1-20140130-C00185
    Compound 205
    Figure US20140030295A1-20140130-C00186
    Compound 206
    Figure US20140030295A1-20140130-C00187
    Compound 207
    Figure US20140030295A1-20140130-C00188
    Compound 208
    Figure US20140030295A1-20140130-C00189
    Compound 209
    Figure US20140030295A1-20140130-C00190
    Compound 210
    Figure US20140030295A1-20140130-C00191
    Compound 211
    Figure US20140030295A1-20140130-C00192
    Compound 212
    Figure US20140030295A1-20140130-C00193
    Compound 213
    Figure US20140030295A1-20140130-C00194
    Compound 214
    Figure US20140030295A1-20140130-C00195
    Compound 215
    Figure US20140030295A1-20140130-C00196
    Compound 216
    Figure US20140030295A1-20140130-C00197
    Compound 217
    Figure US20140030295A1-20140130-C00198
    Compound 218
    Figure US20140030295A1-20140130-C00199
    Compound 219
    Figure US20140030295A1-20140130-C00200
    Compound 220
    Figure US20140030295A1-20140130-C00201
    Compound 221
    Figure US20140030295A1-20140130-C00202
    Compound 222
    Figure US20140030295A1-20140130-C00203
    Compound 223
    Figure US20140030295A1-20140130-C00204
    Compound 225
    Figure US20140030295A1-20140130-C00205
    Compound 226
    Figure US20140030295A1-20140130-C00206
    Compound 227
    Figure US20140030295A1-20140130-C00207
    Compound 228
    Figure US20140030295A1-20140130-C00208
    Compound 229
    Figure US20140030295A1-20140130-C00209
    Compound 230
    Figure US20140030295A1-20140130-C00210
    Compound 231
    Figure US20140030295A1-20140130-C00211
    Compound 232
    Figure US20140030295A1-20140130-C00212
    Compound 234
    Figure US20140030295A1-20140130-C00213
    Compound 235
    Figure US20140030295A1-20140130-C00214
    Compound 236
    Figure US20140030295A1-20140130-C00215
    Compound 237
    Figure US20140030295A1-20140130-C00216
    Compound 238
    Figure US20140030295A1-20140130-C00217
    Compound 239
    Figure US20140030295A1-20140130-C00218
    Compound 240
    Figure US20140030295A1-20140130-C00219
    Compound 241
    Figure US20140030295A1-20140130-C00220
    Compound 244
    Figure US20140030295A1-20140130-C00221
    Compound 245
    Figure US20140030295A1-20140130-C00222
    Compound 246
    Figure US20140030295A1-20140130-C00223
    Compound 247
    Figure US20140030295A1-20140130-C00224
    Compound 248
    Figure US20140030295A1-20140130-C00225
    Compound 249
    Figure US20140030295A1-20140130-C00226
    Compound 250
    Figure US20140030295A1-20140130-C00227
    Compound 251
    Figure US20140030295A1-20140130-C00228
    Compound 252
    Figure US20140030295A1-20140130-C00229
    Compound 253
    Figure US20140030295A1-20140130-C00230
    Compound 254
    Figure US20140030295A1-20140130-C00231
    Compound 255
    Figure US20140030295A1-20140130-C00232
    Compound 256
    Figure US20140030295A1-20140130-C00233
    Compound 257
    Figure US20140030295A1-20140130-C00234
    Compound 258
    Figure US20140030295A1-20140130-C00235
    Compound 259
    Figure US20140030295A1-20140130-C00236
    Compound 260
    Figure US20140030295A1-20140130-C00237
    Compound 261
    Figure US20140030295A1-20140130-C00238
    Compound 262
    Figure US20140030295A1-20140130-C00239
    Compound 263
    Figure US20140030295A1-20140130-C00240
    Compound 264
    Figure US20140030295A1-20140130-C00241
    Compound 265
    Figure US20140030295A1-20140130-C00242
    Compound 266
    Figure US20140030295A1-20140130-C00243
    Compound 267
    Figure US20140030295A1-20140130-C00244
    Compound 268
    Figure US20140030295A1-20140130-C00245
    Compound 270
    Figure US20140030295A1-20140130-C00246
    Compound 271
    Figure US20140030295A1-20140130-C00247
    Compound 272
    Figure US20140030295A1-20140130-C00248
    Compound 273
    Figure US20140030295A1-20140130-C00249
    Compound 276
    Figure US20140030295A1-20140130-C00250
    Compound 280
    Figure US20140030295A1-20140130-C00251
    Compound 282
    Figure US20140030295A1-20140130-C00252
    Compound 283
    Figure US20140030295A1-20140130-C00253
    Compound 284
    Figure US20140030295A1-20140130-C00254
    Compound 285
    Figure US20140030295A1-20140130-C00255
    Compound 286
    Figure US20140030295A1-20140130-C00256
    Compound 287
    Figure US20140030295A1-20140130-C00257
    Compound 288
    Figure US20140030295A1-20140130-C00258
    Compound 289
    Figure US20140030295A1-20140130-C00259
    Compound 290
    Figure US20140030295A1-20140130-C00260
    Compound 292
    Figure US20140030295A1-20140130-C00261
    Compound 293
    Figure US20140030295A1-20140130-C00262
    Compound 294
    Figure US20140030295A1-20140130-C00263
    Compound 295
    Figure US20140030295A1-20140130-C00264
    Compound 296
    Figure US20140030295A1-20140130-C00265
    Compound 297
    Figure US20140030295A1-20140130-C00266
    Compound 298
    Figure US20140030295A1-20140130-C00267
    Compound 299
    Figure US20140030295A1-20140130-C00268
    Compound 303
    Figure US20140030295A1-20140130-C00269
    Compound 304
    Figure US20140030295A1-20140130-C00270
    Compound 305
    Figure US20140030295A1-20140130-C00271
    Compound 306
    Figure US20140030295A1-20140130-C00272
    Compound 307
    Figure US20140030295A1-20140130-C00273
    Compound 308
    Figure US20140030295A1-20140130-C00274
    Compound 309
    Figure US20140030295A1-20140130-C00275
    Compound 310
    Figure US20140030295A1-20140130-C00276
    Compound 311
    Figure US20140030295A1-20140130-C00277
    Compound 313
    Figure US20140030295A1-20140130-C00278
    Compound 314
    Figure US20140030295A1-20140130-C00279
    Compound 315
    Figure US20140030295A1-20140130-C00280
    Compound 317
    Figure US20140030295A1-20140130-C00281
    Compound 318
    Figure US20140030295A1-20140130-C00282
    Compound 319
    Figure US20140030295A1-20140130-C00283
    Compound 320
    Figure US20140030295A1-20140130-C00284
    Compound 321
    Figure US20140030295A1-20140130-C00285
    Compound 322
    Figure US20140030295A1-20140130-C00286
    Compound 323
    Figure US20140030295A1-20140130-C00287
    Compound 324
    Figure US20140030295A1-20140130-C00288
    Compound 325
    Figure US20140030295A1-20140130-C00289
    Compound 326
    Figure US20140030295A1-20140130-C00290
    Compound 327
    Figure US20140030295A1-20140130-C00291
    Compound 328
    Figure US20140030295A1-20140130-C00292
    Compound 329
    Figure US20140030295A1-20140130-C00293
    Compound 330
    Figure US20140030295A1-20140130-C00294
    Compound 331
    Figure US20140030295A1-20140130-C00295
    Compound 332
    Figure US20140030295A1-20140130-C00296
    Compound 333
    Figure US20140030295A1-20140130-C00297
    Compound 334
    Figure US20140030295A1-20140130-C00298
    Compound 335
    Figure US20140030295A1-20140130-C00299
    Compound 336
    Figure US20140030295A1-20140130-C00300
    Compound 337
    Figure US20140030295A1-20140130-C00301
    Compound 338
    Figure US20140030295A1-20140130-C00302
    Compound 339
    Figure US20140030295A1-20140130-C00303
    Compound 340
    Figure US20140030295A1-20140130-C00304
    Compound 341
    Figure US20140030295A1-20140130-C00305
    Compound 342
    Figure US20140030295A1-20140130-C00306
    Compound 343
    Figure US20140030295A1-20140130-C00307
    Compound 344
    Figure US20140030295A1-20140130-C00308
    Compound 345
    Figure US20140030295A1-20140130-C00309
    Compound 346
    Figure US20140030295A1-20140130-C00310
    Compound 347
    Figure US20140030295A1-20140130-C00311
    Compound 348
    Figure US20140030295A1-20140130-C00312
    Compound 349
    Figure US20140030295A1-20140130-C00313
    Compound 350
    Figure US20140030295A1-20140130-C00314
    Compound 351
    Figure US20140030295A1-20140130-C00315
    Compound 359
    Figure US20140030295A1-20140130-C00316
    Compound 362
    Figure US20140030295A1-20140130-C00317
    Compound 364
    Figure US20140030295A1-20140130-C00318
    Compound 367
    Figure US20140030295A1-20140130-C00319
    Compound 369
    Figure US20140030295A1-20140130-C00320
    Compound 370
    Figure US20140030295A1-20140130-C00321
    Compound 371
    Figure US20140030295A1-20140130-C00322
    Compound 372
    Figure US20140030295A1-20140130-C00323
    Compound 373
    Figure US20140030295A1-20140130-C00324
    Compound 374
    Figure US20140030295A1-20140130-C00325
    Compound 375
    Figure US20140030295A1-20140130-C00326
    Compound 376
    Figure US20140030295A1-20140130-C00327
    Compound 377
    Figure US20140030295A1-20140130-C00328
    Compound 378
    Figure US20140030295A1-20140130-C00329
    Compound 379
    Figure US20140030295A1-20140130-C00330
    Compound 380
    Figure US20140030295A1-20140130-C00331
    Compound 381
    Figure US20140030295A1-20140130-C00332
    Compound 382
    Figure US20140030295A1-20140130-C00333
    Compound 383
    Figure US20140030295A1-20140130-C00334
    Compound 384
    Figure US20140030295A1-20140130-C00335
    Compound 385
    Figure US20140030295A1-20140130-C00336
    Compound 387
    Figure US20140030295A1-20140130-C00337
    Compound 390
    Figure US20140030295A1-20140130-C00338
    Compound 391
    Figure US20140030295A1-20140130-C00339
    Compound 392
    Figure US20140030295A1-20140130-C00340
    Compound 393
    Figure US20140030295A1-20140130-C00341
    Compound 394
    Figure US20140030295A1-20140130-C00342
    Compound 396
    Figure US20140030295A1-20140130-C00343
    Compound 398
    Figure US20140030295A1-20140130-C00344
    Compound 399
    Figure US20140030295A1-20140130-C00345
    Compound 400
    Figure US20140030295A1-20140130-C00346
    Compound 401
    Figure US20140030295A1-20140130-C00347
    Compound 402
    Figure US20140030295A1-20140130-C00348
    Compound 403
    Figure US20140030295A1-20140130-C00349
    Compound 404
    Figure US20140030295A1-20140130-C00350
    Compound 405
    Figure US20140030295A1-20140130-C00351
    Compound 406
    Figure US20140030295A1-20140130-C00352
    Compound 407
    Figure US20140030295A1-20140130-C00353
    Compound 408
    Figure US20140030295A1-20140130-C00354
    Compound 409
    Figure US20140030295A1-20140130-C00355
    Compound 410
    Figure US20140030295A1-20140130-C00356
    Compound 411
    Figure US20140030295A1-20140130-C00357
    Compound 412
    Figure US20140030295A1-20140130-C00358
    Compound 413
    Figure US20140030295A1-20140130-C00359
    Compound 414
    Figure US20140030295A1-20140130-C00360
    Compound 415
    Figure US20140030295A1-20140130-C00361
    Compound 416
    Figure US20140030295A1-20140130-C00362
    Compound 419
    Figure US20140030295A1-20140130-C00363
    Compound 420
    Figure US20140030295A1-20140130-C00364
    Compound 421
    Figure US20140030295A1-20140130-C00365
    Compound 422
    Figure US20140030295A1-20140130-C00366
    Compound 423
    Figure US20140030295A1-20140130-C00367
    Compound 424
    Figure US20140030295A1-20140130-C00368
    Compound 425
    Figure US20140030295A1-20140130-C00369
    Compound 426
    Figure US20140030295A1-20140130-C00370
    Compound 427
    Figure US20140030295A1-20140130-C00371
    Compound 428
    Figure US20140030295A1-20140130-C00372
    Compound 429
    Figure US20140030295A1-20140130-C00373
    Compound 430
    Figure US20140030295A1-20140130-C00374
    Compound 431
    Figure US20140030295A1-20140130-C00375
    Compound 436
    Figure US20140030295A1-20140130-C00376
    Compound 437
    Figure US20140030295A1-20140130-C00377
    Compound 438
    Figure US20140030295A1-20140130-C00378
    Compound 439
    Figure US20140030295A1-20140130-C00379
    Compound 440
    Figure US20140030295A1-20140130-C00380
    Compound 441
    Figure US20140030295A1-20140130-C00381
    Compound 442
    Figure US20140030295A1-20140130-C00382
    Compound 443
    Figure US20140030295A1-20140130-C00383
    Compound 444
    Figure US20140030295A1-20140130-C00384
    Compound 445
    Figure US20140030295A1-20140130-C00385
    Compound 446
    Figure US20140030295A1-20140130-C00386
    Compound 447
    Figure US20140030295A1-20140130-C00387
    Compound 448
    Figure US20140030295A1-20140130-C00388
    Compound 449
    Figure US20140030295A1-20140130-C00389
    Compound 450
    Figure US20140030295A1-20140130-C00390
    Compound 451
    Figure US20140030295A1-20140130-C00391
    Compound 452
    Figure US20140030295A1-20140130-C00392
    Compound 453
    Figure US20140030295A1-20140130-C00393
    Compound 454
    Figure US20140030295A1-20140130-C00394
    Compound 455
    Figure US20140030295A1-20140130-C00395
    Compound 456
    Figure US20140030295A1-20140130-C00396
    Compound 457
    Figure US20140030295A1-20140130-C00397
    Compound 458
    Figure US20140030295A1-20140130-C00398
    Compound 459
    Figure US20140030295A1-20140130-C00399
    Compound 460
    Figure US20140030295A1-20140130-C00400
    Compound 461
    Figure US20140030295A1-20140130-C00401
    Compound 462
    Figure US20140030295A1-20140130-C00402
    Compound 463
    Figure US20140030295A1-20140130-C00403
    Compound 465
    Figure US20140030295A1-20140130-C00404
    Compound 466
    Figure US20140030295A1-20140130-C00405
    Compound 467
    Figure US20140030295A1-20140130-C00406
    Compound 468
    Figure US20140030295A1-20140130-C00407
    Compound 469
    Figure US20140030295A1-20140130-C00408
    Compound 470
    Figure US20140030295A1-20140130-C00409
    Compound 471
    Figure US20140030295A1-20140130-C00410
    Compound 472
    Figure US20140030295A1-20140130-C00411
    Compound 473
    Figure US20140030295A1-20140130-C00412
    Compound 474
    Figure US20140030295A1-20140130-C00413
    Compound 475
    Figure US20140030295A1-20140130-C00414
    Compound 476
    Figure US20140030295A1-20140130-C00415
    Compound 477
    Figure US20140030295A1-20140130-C00416
    Compound 478
    Figure US20140030295A1-20140130-C00417
    Compound 479
    Figure US20140030295A1-20140130-C00418
    Compound 481
    Figure US20140030295A1-20140130-C00419
    Compound 482
    Figure US20140030295A1-20140130-C00420
    Compound 483
    Figure US20140030295A1-20140130-C00421
    Compound 484
    Figure US20140030295A1-20140130-C00422
    Compound 485
    Figure US20140030295A1-20140130-C00423
    Compound 486
    Figure US20140030295A1-20140130-C00424
    Compound 487
    Figure US20140030295A1-20140130-C00425
    Compound 488
    Figure US20140030295A1-20140130-C00426
    Compound 489
    Figure US20140030295A1-20140130-C00427
    Compound 490
    Figure US20140030295A1-20140130-C00428
    Compound 491
    Figure US20140030295A1-20140130-C00429
    Compound 492
    Figure US20140030295A1-20140130-C00430
    Compound 493
    Figure US20140030295A1-20140130-C00431
    Compound 494
    Figure US20140030295A1-20140130-C00432
    Compound 495
    Figure US20140030295A1-20140130-C00433
    Compound 496
    Figure US20140030295A1-20140130-C00434
    Compound 497
    Figure US20140030295A1-20140130-C00435
    Compound 498
    Figure US20140030295A1-20140130-C00436
    Compound 499
    Figure US20140030295A1-20140130-C00437
    Compound 500
    Figure US20140030295A1-20140130-C00438
    Compound 501
    Figure US20140030295A1-20140130-C00439
    Compound 502
    Figure US20140030295A1-20140130-C00440
    Compound 503
    Figure US20140030295A1-20140130-C00441
    Compound 504
    Figure US20140030295A1-20140130-C00442
    Compound 505
    Figure US20140030295A1-20140130-C00443
    Compound 506
    Figure US20140030295A1-20140130-C00444
    Compound 507
    Figure US20140030295A1-20140130-C00445
    Compound 508
    Figure US20140030295A1-20140130-C00446
    Compound 510
    Figure US20140030295A1-20140130-C00447
    Compound 511
    Figure US20140030295A1-20140130-C00448
    Compound 512
    Figure US20140030295A1-20140130-C00449
    Compound 513
    Figure US20140030295A1-20140130-C00450
    Compound 514
    Figure US20140030295A1-20140130-C00451
    Compound 515
    Figure US20140030295A1-20140130-C00452
    Compound 516
    Figure US20140030295A1-20140130-C00453
    Compound 517
    Figure US20140030295A1-20140130-C00454
    Compound 518
    Figure US20140030295A1-20140130-C00455
    Compound 519
    Figure US20140030295A1-20140130-C00456
    Compound 520
    Figure US20140030295A1-20140130-C00457
    Compound 521
    Figure US20140030295A1-20140130-C00458
    Compound 522
    Figure US20140030295A1-20140130-C00459
    Compound 523
    Figure US20140030295A1-20140130-C00460
    Compound 524
    Figure US20140030295A1-20140130-C00461
    Compound 525
    Figure US20140030295A1-20140130-C00462
    Compound 526
    Figure US20140030295A1-20140130-C00463
    Compound 527
    Figure US20140030295A1-20140130-C00464
    Compound 528
    Figure US20140030295A1-20140130-C00465
    Compound 529
    Figure US20140030295A1-20140130-C00466
    Compound 530
    Figure US20140030295A1-20140130-C00467
    Compound 531
    Figure US20140030295A1-20140130-C00468
    Compound 532
    Figure US20140030295A1-20140130-C00469
    Compound 533
    Figure US20140030295A1-20140130-C00470
    Compound 534
    Figure US20140030295A1-20140130-C00471
    Compound 535
    Figure US20140030295A1-20140130-C00472
    Compound 536
    Figure US20140030295A1-20140130-C00473
    Compound 537
    Figure US20140030295A1-20140130-C00474
    Compound 538
    Figure US20140030295A1-20140130-C00475
    Compound 539
    Figure US20140030295A1-20140130-C00476
    Compound 540
    Figure US20140030295A1-20140130-C00477
    Compound 541
    Figure US20140030295A1-20140130-C00478
    Compound 542
    Figure US20140030295A1-20140130-C00479
    Compound 543
    Figure US20140030295A1-20140130-C00480
    Compound 544
    Figure US20140030295A1-20140130-C00481
    Compound 545
    Figure US20140030295A1-20140130-C00482
    Compound 546
    Figure US20140030295A1-20140130-C00483
    Compound 547
    Figure US20140030295A1-20140130-C00484
    Compound 548
    Figure US20140030295A1-20140130-C00485
    Compound 556
    Figure US20140030295A1-20140130-C00486
    Compound 557
    Figure US20140030295A1-20140130-C00487
    Compound 558
    Figure US20140030295A1-20140130-C00488
    Compound 559
    Figure US20140030295A1-20140130-C00489
    Compound 560
    Figure US20140030295A1-20140130-C00490
    Compound 561
    Figure US20140030295A1-20140130-C00491
    Compound 562
    Figure US20140030295A1-20140130-C00492
    Compound 563
    Figure US20140030295A1-20140130-C00493
    Compound 565
    Figure US20140030295A1-20140130-C00494
    Compound 566
    Figure US20140030295A1-20140130-C00495
    Compound 567
    Figure US20140030295A1-20140130-C00496
    Compound 568
    Figure US20140030295A1-20140130-C00497
    Compound 569
    Figure US20140030295A1-20140130-C00498
    Compound 570
    Figure US20140030295A1-20140130-C00499
    Compound 571
    Figure US20140030295A1-20140130-C00500
    Compound 572
    Figure US20140030295A1-20140130-C00501
    Compound 573
    Figure US20140030295A1-20140130-C00502
    Compound 574
    Figure US20140030295A1-20140130-C00503
    Compound 575
    Figure US20140030295A1-20140130-C00504
    Compound 576
    Figure US20140030295A1-20140130-C00505
    Compound 577
    Figure US20140030295A1-20140130-C00506
    Compound 578
    Figure US20140030295A1-20140130-C00507
    Compound 579
    Figure US20140030295A1-20140130-C00508
    Compound 580
    Figure US20140030295A1-20140130-C00509
    Compound 581
    Figure US20140030295A1-20140130-C00510
    Compound 582
    Figure US20140030295A1-20140130-C00511
    Compound 583
    Figure US20140030295A1-20140130-C00512
    Compound 584
    Figure US20140030295A1-20140130-C00513
    Compound 585
    Figure US20140030295A1-20140130-C00514
    Compound 587
    Figure US20140030295A1-20140130-C00515
    Compound 588
    Figure US20140030295A1-20140130-C00516
    Compound 589
    Figure US20140030295A1-20140130-C00517
    Compound 590
    Figure US20140030295A1-20140130-C00518
    Compound 591
    Figure US20140030295A1-20140130-C00519
    Compound 592
    Figure US20140030295A1-20140130-C00520
    Compound 593
    Figure US20140030295A1-20140130-C00521
    Compound 594
    Figure US20140030295A1-20140130-C00522
    Compound 596
    Figure US20140030295A1-20140130-C00523
    Compound 597
    Figure US20140030295A1-20140130-C00524
    Compound 599
    Figure US20140030295A1-20140130-C00525
    Compound 600
    Figure US20140030295A1-20140130-C00526
    Compound 601
    Figure US20140030295A1-20140130-C00527
    Compound 604
    Figure US20140030295A1-20140130-C00528
    Compound 605
    Figure US20140030295A1-20140130-C00529
    Compound 607
    Figure US20140030295A1-20140130-C00530
    Compound 608
    Figure US20140030295A1-20140130-C00531
    Compound 609
    Figure US20140030295A1-20140130-C00532
    Compound 610
    Figure US20140030295A1-20140130-C00533
    Compound 611
    Figure US20140030295A1-20140130-C00534
    Compound 612
    Figure US20140030295A1-20140130-C00535
    Compound 614
    Figure US20140030295A1-20140130-C00536
    Compound 615
    Figure US20140030295A1-20140130-C00537
    Compound 616
    Figure US20140030295A1-20140130-C00538
    Compound 617
    Figure US20140030295A1-20140130-C00539
    Compound 618
    Figure US20140030295A1-20140130-C00540
    Compound 619
    Figure US20140030295A1-20140130-C00541
    Compound 620
    Figure US20140030295A1-20140130-C00542
    Compound 621
    Figure US20140030295A1-20140130-C00543
    Compound 622
    Figure US20140030295A1-20140130-C00544
    Compound 623
    Figure US20140030295A1-20140130-C00545
    Compound 624
    Figure US20140030295A1-20140130-C00546
    Compound 625
    Figure US20140030295A1-20140130-C00547
    Compound 628
    Figure US20140030295A1-20140130-C00548
    Compound 629
    Figure US20140030295A1-20140130-C00549
    Compound 630
    Figure US20140030295A1-20140130-C00550
    Compound 631
    Figure US20140030295A1-20140130-C00551
    Compound 632
    Figure US20140030295A1-20140130-C00552
    Compound 633
    Figure US20140030295A1-20140130-C00553
    Compound 634
    Figure US20140030295A1-20140130-C00554
    Compound 635
    Figure US20140030295A1-20140130-C00555
    Compound 636
    Figure US20140030295A1-20140130-C00556
    Compound 637
    Figure US20140030295A1-20140130-C00557
    Compound 638
    Figure US20140030295A1-20140130-C00558
    Compound 639
    Figure US20140030295A1-20140130-C00559
    Compound 640
    Figure US20140030295A1-20140130-C00560
    Compound 641
    Figure US20140030295A1-20140130-C00561
    Compound 642
    Figure US20140030295A1-20140130-C00562
    Compound 643
    Figure US20140030295A1-20140130-C00563
    Compound 644
    Figure US20140030295A1-20140130-C00564
    Compound 645
    Figure US20140030295A1-20140130-C00565
    Compound 646
    Figure US20140030295A1-20140130-C00566
    Compound 647
    Figure US20140030295A1-20140130-C00567
    Compound 648
    Figure US20140030295A1-20140130-C00568
    Compound 649
    Figure US20140030295A1-20140130-C00569
    Compound 650
    Figure US20140030295A1-20140130-C00570
    Compound 651
    Figure US20140030295A1-20140130-C00571
    Compound 655
    Figure US20140030295A1-20140130-C00572
    Compound 656
    Figure US20140030295A1-20140130-C00573
    Compound 657
    Figure US20140030295A1-20140130-C00574
    Compound 658
    Figure US20140030295A1-20140130-C00575
    Compound 659
    Figure US20140030295A1-20140130-C00576
    Compound 660
    Figure US20140030295A1-20140130-C00577
    Compound 661
    Figure US20140030295A1-20140130-C00578
    Compound 662
    Figure US20140030295A1-20140130-C00579
    Compound 663
    Figure US20140030295A1-20140130-C00580
    Compound 664
    Figure US20140030295A1-20140130-C00581
    Compound 665
    Figure US20140030295A1-20140130-C00582
    Compound 666
    Figure US20140030295A1-20140130-C00583
    Compound 667
    Figure US20140030295A1-20140130-C00584
    Compound 668
    Figure US20140030295A1-20140130-C00585
    Compound 669
    Figure US20140030295A1-20140130-C00586
    Compound 670
    Figure US20140030295A1-20140130-C00587
    Compound 671
    Figure US20140030295A1-20140130-C00588
    Compound 672
    Figure US20140030295A1-20140130-C00589
    Compound 673
    Figure US20140030295A1-20140130-C00590
    Compound 674
    Figure US20140030295A1-20140130-C00591
    Compound 675
    Figure US20140030295A1-20140130-C00592
    Compound 676
    Figure US20140030295A1-20140130-C00593
    Compound 677
    Figure US20140030295A1-20140130-C00594
    Compound 678
    Figure US20140030295A1-20140130-C00595
    Compound 679
    Figure US20140030295A1-20140130-C00596
    Compound 680
    Figure US20140030295A1-20140130-C00597
    Compound 681
    Figure US20140030295A1-20140130-C00598
    Compound 682
    Figure US20140030295A1-20140130-C00599
    Compound 683
    Figure US20140030295A1-20140130-C00600
    Compound 684
    Figure US20140030295A1-20140130-C00601
    Compound 685
    Figure US20140030295A1-20140130-C00602
    Compound 686
    Figure US20140030295A1-20140130-C00603
    Compound 687
    Figure US20140030295A1-20140130-C00604
    Compound 688
    Figure US20140030295A1-20140130-C00605
    Compound 689
    Figure US20140030295A1-20140130-C00606
    Compound 690
    Figure US20140030295A1-20140130-C00607
    Compound 692
    Figure US20140030295A1-20140130-C00608
    Compound 695
    Figure US20140030295A1-20140130-C00609
    Compound 697
    Figure US20140030295A1-20140130-C00610
    Compound 698
    Figure US20140030295A1-20140130-C00611
    Compound 699
    Figure US20140030295A1-20140130-C00612
    Compound 700
    Figure US20140030295A1-20140130-C00613
    Compound 701
    Figure US20140030295A1-20140130-C00614
    Compound 702
    Figure US20140030295A1-20140130-C00615
    Compound 703
    Figure US20140030295A1-20140130-C00616
    Compound 704
    Figure US20140030295A1-20140130-C00617
    Compound 705
    Figure US20140030295A1-20140130-C00618
    Compound 706
    Figure US20140030295A1-20140130-C00619
    Compound 707
    Figure US20140030295A1-20140130-C00620
    Compound 708
    Figure US20140030295A1-20140130-C00621
    Compound 709
    Figure US20140030295A1-20140130-C00622
    Compound 710
    Figure US20140030295A1-20140130-C00623
    Compound 711
    Figure US20140030295A1-20140130-C00624
    Compound 714
    Figure US20140030295A1-20140130-C00625
    Compound 715
    Figure US20140030295A1-20140130-C00626
    Compound 716
    Figure US20140030295A1-20140130-C00627
    Compound 717
    Figure US20140030295A1-20140130-C00628
    Compound 718
    Figure US20140030295A1-20140130-C00629
    Compound 719
    Figure US20140030295A1-20140130-C00630
    Compound 720
    Figure US20140030295A1-20140130-C00631
    Compound 721
    Figure US20140030295A1-20140130-C00632
    Compound 722
    Figure US20140030295A1-20140130-C00633
    Compound 723
    Figure US20140030295A1-20140130-C00634
    Compound 724
    Figure US20140030295A1-20140130-C00635
    Compound 725
    Figure US20140030295A1-20140130-C00636
    Compound 726
    Figure US20140030295A1-20140130-C00637
    Compound 727
    Figure US20140030295A1-20140130-C00638
    Compound 728
    Figure US20140030295A1-20140130-C00639
    Compound 729
    Figure US20140030295A1-20140130-C00640
    Compound 730
    Figure US20140030295A1-20140130-C00641
    Compound 731
    Figure US20140030295A1-20140130-C00642
    Compound 732
    Figure US20140030295A1-20140130-C00643
    Compound 733
    Figure US20140030295A1-20140130-C00644
    Compound 735
    Figure US20140030295A1-20140130-C00645
    Compound 736
    Figure US20140030295A1-20140130-C00646
    Compound 737
    Figure US20140030295A1-20140130-C00647
    Compound 738
    Figure US20140030295A1-20140130-C00648
    Compound 739
    Figure US20140030295A1-20140130-C00649
    Compound 740
    Figure US20140030295A1-20140130-C00650
    Compound 741
    Figure US20140030295A1-20140130-C00651
    Compound 742
    Figure US20140030295A1-20140130-C00652
    Compound 743
    Figure US20140030295A1-20140130-C00653
    Compound 744
    Figure US20140030295A1-20140130-C00654
    Compound 745
  • Non-limiting examples of suitable SIRT1 activators include, e.g.,
  • Figure US20140030295A1-20140130-C00655
  • Other suitable SIRT1 activators include, e.g.,
  • Figure US20140030295A1-20140130-C00656
  • SRT1720, SRT1460, and SRT2183 are selective SIRT1 activators. See, e.g., Milne et al. (2007) Nature 450:712.
  • Also suitable for use are SIRT1 activators that are quinoxaline compounds. Suitable quinoxaline SIRT1 activators include, e.g., 3-benzenesulfonyl-1-(4-fluoro-phenyl)-1H-pyrrolo[2,3-b]quinoxalin-2-ylamine; 2-amino-1-(2-ethyl-phenyl)-1H-pyrrolo[2,3-b]quinoxaline-3-carboxylic acid (tetrahydro-furan-2-ylmethyl)-amine; 2-amino-1-(3-methoxy-propyl)-1H-pyrrolo[2,3-b]quinoxaline-3-carboxylic acid cyclopentylamide. See, e.g., Nayagam et al. (2006) J. Biolmolec. Screening 11:959.
  • Other suitable SIRT1 activators include, e.g., stilbene compounds, e.g., ester analogs of resveratrol, e.g., as described in U.S. Patent Publication No. 2008/0255382. For example, suitable SIRT1 activators include, e.g., ester analogs of 3,5,4′-trihydroxy-trans-stilbene.
  • Figure US20140030295A1-20140130-C00657
  • Ester analogs include compounds of the formula:
  • Figure US20140030295A1-20140130-C00658
  • where each Y and each Z is independently —O (ethers), —O—C═O; —C═O—O (esters); —O—C═O—O (carbonates); —O—C═O—NH; —O═O—NR; —NH—C═O—O; —NR—C═O—O (carbamates); —NH—C═P; —NR—C═O; —C═O—NH; —C═O—NR (primary and secondary amides)-NH; —NR (primary and secondary amines); —N (heterocyclic rings); —S (thiol ethers); and halogen;
  • where each n and each m is independently 1, 2, 3, 4, or 5;
  • where each A and each B is independently H, R, or absent;
  • where each V and each W is independently H, straight or branched alkyl of from 1 to 6 carbon atoms, cycloalkyl of from 3 to 8 carbon atoms, alkoxy, phenyl, benzyl, or halogen,
  • and where R is an alkyl with at least one carbon atom, an aryl, or an aralkyl.
  • Suitable SIRT1 activators include, e.g., 4′-acetoxy-3,5-bis(methoxymethoxy)stilbene; 4′-acetoxy-3,5-dihydroxystilbene; 3,5-diacetoxy-4′-chloroacetoxy stilbene; 3,5-diacetoxy-4′-hydroxy stilbene; 3,4′-diacetoxy-5-hydroxystilbene; 3-acetoxy-4′5-dihydroxystilbene; and 3,4,5′-triacetoxystilbene.
  • Suitable SIRT1 activators include compounds of any one of Formulas I-VI as described in U.S. Patent Publication No. 2009/0012080. For example, a suitable SIRT1 activator is a compound of the formula:
  • Figure US20140030295A1-20140130-C00659
  • For example, a suitable SIRT1 activator is 4-methyl-N-(2(3-morpholinomethyl)imidazol[2,1-b]thiazol-6-yl)phenyl)-2-(pyridin-3-yl)thiazol-5-carboxamide, or a pharmaceutically acceptable salt thereof.
  • Methods of Modulating Activation and Differentiation of a CD4+ T Cell
  • The present disclosure provides a method of modulating activation and differentiation of a CD4+ T cell, the method generally involving contacting the CD4+ T cell (in vitro, ex vivo, or in vivo) with a SIRT1 inhibitor. In some embodiments, the CD4+ T cell is a helper T cell (e.g., a Th1, Th2, or Th17 cell), and the SIRT1 inhibitor inhibits deacetylation of NF-κB in the CD4+ T cell, thereby increasing CD4+ T helper T cell activity and/or numbers. In other embodiments, the CD4+ T cell is a CD4+/CD25+/FoxP3+ Treg cell, and the SIRT1 inhibitor reduces deacetylation of FoxP3 in the CD4+/CD25+/FoxP3+ Treg cell, thereby increasing Treg activity and/or numbers.
  • Increasing CD4+ T Helper Cell Activity and/or Numbers
  • In some embodiments, the CD4+ T cell is a helper T cell (e.g., a Th1, a Th2, or a Th17 cell), and the SIRT1 inhibitor inhibits deacetylation of NF-κB in the CD4+ T cell, thereby increasing CD4+ T helper cell activity and/or numbers. CD4+ T helper cell activity and/or numbers can be increased in vitro, in vivo, or ex vivo. Increasing CD4+ T helper cell activity and/or numbers is useful for increasing an immune response, e.g., increasing an immune response to an antigen. Thus, the present disclosure provides a method of increasing an immune response in an individual, the method involving administering to an individual in need thereof an effective amount of an inhibitor of SIRT1. The method can also be carried out ex vivo, e.g., by (a) contacting CD4+ T helper cells obtained from a donor individual with a SIRT1 inhibitor ex vivo, thereby increasing the activity and/or numbers of CD4+ T helper cells, and (b) returning the CD4+ T helper cells from step (a) to the donor individual or to a recipient individual other than the donor.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that, when contacted with a starting population of CD4+ T helper cells, increases the number of CD4+ T helper cells by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold, or more, compared to the number of CD4+ T helper cells in the starting population before contacting with the SIRT1 inhibitor. As noted above, in some embodiments, the contacting occurs in vitro. In other embodiments, the contacting occurs ex vivo. In other embodiments, the contacting occurs in vivo.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce deacetylation of NF-κB in a CD4+ T helper cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of deacetylation of NF-κB in the CD4+ T helper cell in the absence of the SIRT1 inhibitor.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that, when administered to an individual, increases an immune response in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the immune response in the absence of treatment with the SIRT1 inhibitor.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that increases the number of CD4+ cells in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the number of CD4+ T cells in the individual in the absence of treatment with the SIRT1 inhibitor.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that increases the number of cytotoxic T cells in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the number of cytotoxic T cells in the individual in the absence of treatment with the SIRT1 inhibitor.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce the number of cancer cells and/or to reduce the volume of a tumor in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the number of cancer cells or compared to the tumor volume in the individual in the absence of treatment with the SIRT1 inhibitor. In some embodiment, a SIRT1 inhibitor is administered in conjunction with administration of a tumor-specific antigen.
  • In some embodiments, a SIRT1 inhibitor is administered in conjunction with administration of an antigen that induces an immune response to a pathological microorganism. In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce the number of pathological microorganisms in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the number of pathological microorganisms in the individual in the absence of treatment with the SIRT1 inhibitor. In some embodiment, a SIRT1 inhibitor is administered in conjunction with administration of an antigen that induces an immune response to a pathological microorganism.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that increases the level of one or more antigen-specific antibodies in an individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, or at least about 100-fold, compared to the level of the one or more antigen-specific antibodies in the individual in the absence of treatment with the SIRT1 inhibitor.
  • Analysis (both qualitative and quantitative) an immune response can be by any method known in the art, including, but not limited to, measuring antigen-specific antibody production, activation of specific populations of lymphocytes such as CD4+ T cells or NK cells, and/or production of cytokines such as IFN, IL-2, IL-4, or IL-12. Methods for measuring specific antibody responses include enzyme-linked immunosorbent assay (ELISA) and are well known in the art. Measurement of numbers of specific types of lymphocytes such as CD4+ T cells can be achieved, for example, with fluorescence-activated cell sorting (FACS). Cytotoxicity assays can be performed for instance as described in Raz et al. (1994) Proc. Natl. Acad. Sci. USA 91: 9519-9523. Serum concentrations of cytokines can be measured, for example, by ELISA. These and other assays to evaluate the immune response are well known in the art. In some embodiments, a Th1-type immune response is increased.
  • A subject method of increasing an immune response in an individual is useful for treating various disorders including, e.g., treating an infectious disease (e.g., a disease caused by a pathological microorganism such as a virus, a bacterium, etc.); increasing an immune response to a cancer cell in an individual; enhancing an immune response in an individual who is immunodeficient or immunocompromised; etc.
  • Thus, for example, a subject method can be used to increase an immune response for treating:
  • (a) viral diseases such as, for example, diseases resulting from infection by an adenovirus, a herpesvirus (e.g., HSV-I, HSV-II, CMV, or VZV), a poxvirus (e.g., an orthopoxvirus such as variola or vaccinia), a picornavirus (e.g., rhinovirus or enterovirus), an orthomyxovirus (e.g., influenzavirus), a paramyxovirus (e.g., parainfluenzavirus, mumps virus, measles virus, and respiratory syncytial virus (RSV)), a coronavirus (e.g., SARS), a papovavirus (e.g., papillomaviruses, such as those that cause genital warts, common warts, or plantar warts), a hepadnavirus (e.g., hepatitis B virus), a flavivirus (e.g., hepatitis C virus or Dengue virus), or a retrovirus (e.g., a lentivirus such as HIV);
  • (b) bacterial diseases such as, for example, diseases resulting from infection by bacteria of, for example, the genus Escherichia, Enterobacter, Salmonella, Staphylococcus, Shigella, Listeria, Aerobacter, Helicobacter, Klebsiella, Proteus, Pseudomonas, Streptococcus, Chlamydia, Mycoplasma, Pneumococcus, Neisseria, Clostridium, Bacillus, Corynebacterium, Mycobacterium, Campylobacter, Vibrio, Serratia, Providencia, Chromobacterium, Brucella, Yersinia, Haemophilus, and Bordetella;
  • (c) other infectious diseases, such Chlamydia infection, fungal diseases including but not limited to candidiasis, aspergillosis, histoplasmosis, cryptococcal meningitis, and parasitic diseases including but not limited to malaria, Pneumocystis carinii pneumonia, leishmaniasis, cryptosporidiosis, toxoplasmosis, and trypanosome infection; and
  • (d) neoplastic diseases, such as, for example, intraepithelial neoplasias, cervical dysplasia, actinic keratosis, basal cell carcinoma, squamous cell carcinoma, renal cell carcinoma, Kaposi's sarcoma, melanoma, renal cell carcinoma, leukemias including but not limited to myelogeous leukemia, chronic lymphocytic leukemia, multiple myeloma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, B-cell lymphoma, and hairy cell leukemia, and other cancers.
  • Increasing Treg Activity and/or Numbers
  • In other embodiments, the CD4+ T cell is a CD4+/CD25+/FoxP3+ Treg cell, and the SIRT1 inhibitor reduces deacetylation of FoxP3 in the CD4+/CD25+/FoxP3+ Treg cell, thereby increasing Treg activity and/or numbers. Thus, in some embodiments, the present disclosure provides a method of increasing Treg activity and/or numbers, the method generally involving contacting a naïve CD4+ T cell and/or a Treg cell (a CD4+/CD25+/FoxP3+ T cell) with a SIRT1 modulator, where the contacting can take place in vitro, ex vivo, or in vivo.
  • Increasing Treg activity and/or numbers is useful for reducing an undesirable immune response such as an autoimmune response, an allergic response, graft rejection, or a graft-versus-host response. Thus, in some embodiments, the present disclosure provides a method of treating an autoimmune disorder, a method of treating an allergic disorder, a method of reducing graft rejection, and a method of reducing graft-versus-host disease, in an individual, the methods generally involving administering to an individual in need thereof (e.g., an individual having an autoimmune disorder, an individual having an allergic disorder, a graft recipient) an effective amount of an SIRT1 inhibitor.
  • In some embodiments, an autoimmune response, an allergic response, graft rejection, or a graft-versus-host response, can be reduced by contacting naïve CD4+ T cells and/or Treg cells obtained from an individual (e.g., an individual having an autoimmune disorder, an individual having an allergic disorder, a graft recipient) with an SIRT1 inhibitor ex vivo, and returning the Treg cells to the individual. For example, an ex vivo method can involve: (a) contacting naïve CD4+ T cells and/or Tregs obtained from a donor individual with a SIRT1 inhibitor ex vivo, thereby increasing the activity and/or numbers of Tregs, and (b) returning the Tregs from step (a) to the donor individual or to a recipient individual other than the donor.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that, when contacted with a starting population of Tregs (or a starting population that is a mixture of naïve CD4+ T cells and Tregs), increases the number of Tregs by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, at least about 25-fold, at least about 50-fold, at least about 100-fold, at least about 500-fold, or at least about 1000-fold, or more, compared to the number of Tregs in the starting population before contacting with the SIRT1 inhibitor. As noted above, in some embodiments, the contacting occurs in vitro. In other embodiments, the contacting occurs ex vivo. In other embodiments, the contacting occurs in vivo.
  • In some embodiments, an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce deacetylation of FoxP3 in a Treg by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of deacetylation of FoxP3 in the Treg in the absence of the SIRT1 inhibitor. For example, in some embodiments, an effective amount of a SIRT1 inhibitor is an amount that is effective to reduce deacetylation of K31, K263, and K268 (or an equivalent lysine residue) in a FoxP3 polypeptide in a Treg cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of deacetylation of K31, K263, and K268 residues in FoxP3 in the Treg in the absence of the SIRT1 inhibitor.
  • Amino acid sequences of FoxP3 polypeptides are known in the art. Examples of FoxP3 amino acid sequences are presented in FIG. 13, which shows an alignment of human, mouse, and bovine FoxP3, and which shows the K31, K263, and K268 residues. A FoxP3 polypeptide can comprise an amino acid sequence having at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, with the human FoxP3 amino acid sequence depicted in FIG. 13.
  • In some embodiments, an “effective amount” of a SIRT1 inhibitor is an amount that achieves a degree of immunosuppression sufficient to delay, inhibit, suppress or moderate tissue transplant rejection and/or delay, inhibit, suppress or moderate one or more symptoms of an autoimmune disease and/or delay, inhibit, suppress or moderate an undesired immune response to a foreign antigen such as a therapeutic protein, a viral vector, an allergen, a venom and the like.
  • In some embodiments, an “effective amount” of a SIRT1 inhibitor is an amount that reduces an undesired immune response (e.g., an autoimmune response, an allergic response, a graft-versus-host response, a graft rejection) by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the degree of the undesired immune response in the absence of treatment with the SIRT1 inhibitor. Whether an SIRT1 inhibitor is effective in reducing an undesired immune response (e.g., an autoimmune response, an allergic response, a graft-versus-host response, a graft rejection) can be determined by measuring one or more well-known parameters associated with the undesired immune response (e.g., the level of autoantibody; the level of auto-reactive T cells; the level of allergen-specific IgE; a symptom of allergy; the level of antibody specific for a graft tissue; etc.).
  • In some embodiments, a subject method is effective to reduce the number and/or activity of an autoreactive cell in an individual by at least about 5%, at least about 10%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90%, or more, when compared to the number and/or level of autoreactive cells in the individual not treated with a SIRT1 inhibitor.
  • Whether administration of a SIRT1 inhibitor is effective to reduce the number and/or activity of an autoreactive T lymphocyte in an individual is readily determined using known assays. For example, where the autoreactive T lymphocytes are specific for an autoantigen, the number and activity level of autoantigen-specific T lymphocytes is determined using, e.g., a mixed lymphocyte reaction in which irradiated cells comprising a detectable label in the cytoplasm and displaying the autoantigen are mixed with lymphocytes from the individual. Release of detectable label from the cytoplasm of the autoantigen-displaying cells indicates the presence in the individual of autoreactive lymphocytes. Methods of detecting autoreactive T lymphocytes associated with Type 1 diabetes are known in the art; and any such methods can be used. See, e.g., U.S. Pat. No. 6,022,697 for a discussion of a method of detecting autoreactive T lymphocytes associated with Type 1 diabetes.
  • A subject method is useful for treating an autoimmune disease, e.g., for reducing or ameliorating at least one symptom of an autoimmune disease in an individual having an autoimmune disease (e.g., an individual who has been diagnosed as having an autoimmune diseases). Autoimmune diseases include but are not limited to rheumatoid arthritis, Crohn's disease, multiple sclerosis, systemic lupus erythematosus (SLE), autoimmune encephalomyelitis, myasthenia gravis (MG), Hashimoto's thyroiditis, Goodpasture's syndrome, pemphigus (e.g., pemphigus vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis, pernicious anemia, idiopathic Addison's disease, autoimmune-associated infertility, glomerulonephritis (e.g., crescentic glomerulonephritis, proliferative glomerulonephritis), bullous pemphigoid, Sjogren's syndrome, insulin resistance, and autoimmune diabetes mellitus (type 1 diabetes mellitus; insulin-dependent diabetes mellitus).
  • A subject method can be used to increase transplantation tolerance in a subject (e.g., to reduce the likelihood of rejection of the transplant by the transplant recipient). In some embodiments, the subject is a recipient of an allogeneic transplant (e.g., the graft is an allograft). Cells, tissues or organs (or parts thereof) that can be transplanted between members of the same species include, but are not limited, to heart, lung, kidney, liver, pancreas, pancreatic islets, brain tissue, cornea, stomach, bone, bone marrow, muscle, intestine, bladder, skin and stem cells. Optionally, the transplanted tissue or organ is bio-engineered, e.g., when the transplanted tissue or organ is grown from a stem cell or other type of precursor cell(s). Bio-engineered tissue or organ can be grown outside of the body and transplanted directly into the host. Alternatively, the precursor cells or immature organ or tissue is transplanted into the host to grow and mature.
  • The transplanted organ, tissue or cell(s) can also be a xenograft, i.e., the donor is a member of a species different than the recipient. Xenografts are advantageously used with a bio-engineered tissue or organ, which, instead of being transplanted directly into the recipient in need of the tissue or organ, can be transplanted into a surrogate host such as non-human mammal until a suitable human recipient in need of the bio-engineered tissue or organ is identified. Alternatively, the tissue or organ can be transplanted into the surrogate to allow the bio-engineered tissue or organ to mature. Use of surrogate hosts may be preferred in instances where further development of the tissue or organ is required before transplantation into a human recipient. In another alternative, a xenograft is used when a suitable allograft donor is unavailable. When a transplanting into a different a species, it is desirable to select a host such that the size of the organs in the host and donor are similar. In addition, the host is selected to minimize transmission of communicable diseases.
  • In some embodiments, a subject method can be used to reduce the incidence and/or severity of an allergic reaction in an individual. Thus, a subject method can be used to reduce adverse reaction in an individual to an allergen. Allergens of interest include antigens found in food, such as strawberries, peanuts, milk polypeptides, egg whites, etc. Other allergens of interest include various airborne antigens, such as grass pollens, animal danders, house mite feces, etc. Molecularly cloned allergens include Dermatophagoides pteryonyssinus (Der P1); Lol pl-V from rye grass pollen; a number of insect venoms, including venom from jumper ant Myrmecia pilosula; Apis mellifera bee venom phospholipase A2 (PLA2 and antigen 5S; phospholipases from the yellow jacket Vespula maculifrons and white faced hornet Dolichovespula maculata; a large number of pollen polypeptides, including birch pollen, ragweed pollen, Parol (the major allergen of Parietaria officinalis) and the cross-reactive allergen Parjl (from Parietaria judaica), and other atmospheric pollens including Olea europaea, Artemisia sp., gramineae, etc. Other allergens of interest are those responsible for allergic dermatitis caused by blood sucking arthropods, e.g. Diptera, including mosquitos (Anopheles sp., Aedes sp., Culiseta sp., Culex sp.); flies (Phlebotomus sp., Culicoides sp.) particularly black flies, deer flies and biting midges; ticks (Dermacenter sp., Ornithodoros sp., Otobius sp.); fleas, e.g. the order Siphonaptera, including the genera Xenopsylla, Pulex and Ctenocephalides felis. Other allergens of interest include drug allergens.
  • SIRT1 Inhibitors
  • SIRT1 inhibitors that are suitable for use in a subject method include, but are not limited to, sirtinol (2-[(2-hydroxynaphthalen-1-ylmethylene)amino]-N-(1-phenyl-ethyl)benzamide); a compound as described in U.S. Pat. No. 7,345,178, provided the compound is an SIRT1 inhibitor, e.g., a compound as shown in Table 5 of U.S. Pat. No. 7,345,178; a compound as described in U.S. Patent Publication No. 2008/0255382; and the like; or a pharmaceutically acceptable salt of any of the foregoing compounds.
  • Suitable SIRT1 inhibitors include, e.g.,
  • Figure US20140030295A1-20140130-C00660
  • Ex-527 is 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide. Ex-527 is a selective SIRT1 inhibitor. See, e.g., Napper et al. (2005) J. Med. Chem. 48:8045.
  • Suitable SIRT1 inhibitors include, e.g., compounds of Table 5 of U.S. Pat. No. 7,345,178, e.g.:
  • Figure US20140030295A1-20140130-C00661
    Figure US20140030295A1-20140130-C00662
    Figure US20140030295A1-20140130-C00663
  • Suitable SIRT1 inhibitors include a compound as disclosed in U.S. Patent Publication No. 20008/0214800, or a pharmaceutically acceptable salt of such a compound. Examples of suitable SIRT1 inhibitors include, e.g.:
  • Figure US20140030295A1-20140130-C00664
  • Suitable SIRT1 inhibitors include, e.g., 7-chloro-1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; 6-bromo-2,3,4,9-tetrahydro-1H-carbazole-2-carboxylic acid amide; 1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; and 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid (5-chloro-pyridin-2-yl)-amide.
  • Combination Therapy
  • In some embodiments, a subject treatment method comprises administering to an individual in need thereof an effective amount of a SIRT1 inhibitor or a SIRT1 activator, and at least one additional therapeutic agent.
  • HIV-Associated Immune Hyperactivation
  • In some embodiments, as discussed above, a subject method provides for treating immune hyperactivation that is associated with an immunodeficiency virus infection. In some embodiments, a subject method for reducing immunodeficiency virus infection-associated immune hyperactivation involves administering a SIRT1 activator alone, e.g., as monotherapy. In other embodiments, a subject method for reducing immunodeficiency virus infection-associated immune hyperactivation involves administering a SIRT1 activator, and at least one additional therapeutic agent, in combination therapy.
  • Suitable additional therapeutic agents include, e.g., therapeutic agents for the treatment of an immunodeficiency virus infection, or for the treatment of a disorder that may accompany an immunodeficiency virus infection (e.g., a bacterial infection, a fungal infection, and the like). Suitable additional therapeutic agents include, e.g., beta-lactam antibiotics, tetracyclines, chloramphenicol, neomycin, gramicidin, bacitracin, sulfonamides, nitrofurazone, nalidixic acid, cortisone, hydrocortisone, betamethasone, dexamethasone, fluocortolone, prednisolone, triamcinolone, indomethacin, sulindac, acyclovir, amantadine, rimantadine, recombinant soluble CD4 (rsCD4), anti-receptor antibodies (e.g., for rhinoviruses), nevirapine, cidofovir (Vistide™), trisodium phosphonoformate (Foscarnet™), famcyclovir, pencyclovir, valacyclovir, nucleic acid/replication inhibitors, interferon, zidovudine (AZT, Retrovir™) didanosine (dideoxyinosine, ddI, Videx™), stavudine (d4T, Zerit™), zalcitabine (dideoxycytosine, ddC, Hivid™), nevirapine (Viramune™), lamivudine (Epivir™, 3TC), protease inhibitors, saquinavir (Invirase™, Fortovase™), ritonavir (Norvir™), nelfinavir (Viracept™), efavirenz (Sustiva™), abacavir (Ziagen™), amprenavir (Agenerase™) indinavir (Crixivan™), ganciclovir, AzDU, delavirdine (Rescriptor™), kaletra, trizivir, rifampin, clathiromycin, erythropoietin, colony stimulating factors (G-CSF and GM-CSF), non-nucleoside reverse transcriptase inhibitors, nucleoside inhibitors, adriamycin, fluorouracil, methotrexate, asparaginase and combinations thereof.
  • Increasing an Immune Response
  • In some embodiments, a subject method of increasing an immune response comprises administering to an individual in need thereof a SIRT1 inhibitor and at least one additional therapeutic agent in combined effective amounts to increase an immune response in the individual. In some embodiments, a subject method of increasing an immune response comprises administering to an individual in need thereof a SIRT1 inhibitor and an antigen in combined effective amounts to increase an immune response to the antigen in the individual. In some embodiments, a subject method of increasing an immune response comprises administering to an individual in need thereof a SIRT1 inhibitor and at least one additional therapeutic agent in combined effective amounts to increase an immune response in the individual; and administering an antigen to the individual, where an immune response to the antigen is increased.
  • Suitable additional therapeutic agents include, e.g., immunostimulatory polynucleotides (“ISS”; see, e.g., U.S. Pat. No. 7,479,285); an ISS conjugated to an antigen (see, e.g., U.S. Pat. No. 6,610,661); a Toll-like receptor agonist (see, e.g., U.S. Pat. No. 7,387,271); an immunostimulatory cytokine (e.g., IL-2); and the like. Suitable ISS include polynucleotides that include an unmethylated 5′-CG-3′ sequence. ISS can be from about 6 nucleotides in length to about 200 nucleotides in length, or longer than 200 nucleotides in length. An ISS can comprise a phosphate backbone modification, where backbone phosphate group modifications include, e.g., methylphosphonate, phosphorothioate, phosphoroamidate and phosphorodithioate internucleotide linkages.
  • TLR agonists include, e.g., TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8 and TLR9 agonists. Suitable TLR agonists include, e.g., an imidazoquinoline amine, a tetrahydroimidazoquinoline amine, an imidazopyridine amine, a 1,2-bridged imidazoquinoline amine, a 6,7-fused cycloalkylimidazopyridine amine, an imidazonaphthyridine amine, a tetrahydroimidazonaphthyridine amine, an oxazoloquinoline amine, a thiazoloquinoline amine, an oxazolopyridine amine, a thiazolopyridine amine, an oxazolonaphthyridine amine, and a thiazolonaphthyridine amine.
  • Reducing an Undesired Immune Response
  • In some embodiments, a subject method of reducing an undesired immune response (e.g., an autoimmune response; an allergic response; a graft-versus-host response; rejection of a transplanted organ, tissue, or cell) involves administering a SIRT1 inhibitor alone, e.g., as monotherapy. In other embodiments, a subject method of reducing an undesired immune response (e.g., an autoimmune response; an allergic response; a graft-versus-host response; rejection of a transplanted organ, tissue, or cell) involves administering a SIRT1 inhibitor in combination therapy with one or more additional therapeutic agents.
  • For the treatment of autoimmune disorders, a SIRT1 inhibitor can be administered in combination therapy with one or more agents for treating an autoimmune disorder. Those skilled in the art are aware of agents that are suitable for treating autoimmune disorders. For example, agents that are suitable for treating Type 1 diabetes include insulin, including naturally occurring insulin, insulin analogs, and the like. Interferon-alpha is an agent that is currently in use for treating multiple sclerosis, and can be used in combination therapy with a SIRT1 inhibitor. Other agents that are currently in use for treating autoimmune disorders include corticosteroid drugs; non-steroidal anti-inflammatory drugs (NSAIDs); and immunosuppressant drugs such as cyclophosphamide, methotrexate, and azathioprine.
  • For the treatment of allergies, a SIRT1 inhibitor can be administered in combination therapy with one or more agents for treating an allergic disorder. Suitable agents include, but are not limited to, antihistamines such as diphenhydramine (Benadryl); epinephrine; decongestants; and the like.
  • For reduction of graft rejection or for reduction of GVHD, a SIRT1 inhibitor can be administered in combination therapy with one or more agents for reducing graft rejection or GVHD. Agents suitable for reducing graft rejection or GVHD include, but are not limited to, glucocorticoids (e.g., cortisol; dexamethasone; etc.); cytostatic agents (e.g., cyclophosphamide; nitrosoureas; platinum compounds; methotrexate; azathioprine; mercaptopurine; dactinomycin; anthracyclines; mitomycin C; bleomycin; mitramycin; etc.); an anti-CD20 monoclonal antibody; cyclosporin; tacrolimus; voclosporin; sirolimus; and the like.
  • Formulations, Dosages, Routes of Administration
  • A SIRT1 activator or a SIRT1 inhibitor can be formulated with one or more pharmaceutically acceptable excipients. SIRT1 activators and SIRT1 inhibitors are referred to collectively below as “active agent” or “drug.” A wide variety of pharmaceutically acceptable excipients are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, A. Gennaro (2000) “Remington: The Science and Practice of Pharmacy,” 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H. C. Ansel et al., eds., 7th ed., Lippincott, Williams, & Wilkins; and Handbook of Pharmaceutical Excipients (2000) A. H. Kibbe et al., eds., 3rd ed. Amer. Pharmaceutical Assoc.
  • The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • The term “unit dosage form,” as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of a subject active agent calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for an active agent depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • In the subject methods, a SIRT1 activator or a SIRT1 inhibitor may be administered to the host using any convenient means capable of resulting in the desired outcome, e.g., reduction of disease, reduction of a symptom of a disease, etc. Thus, a SIRT1 activator or a SIRT1 inhibitor can be incorporated into a variety of formulations for therapeutic administration. More particularly, a SIRT1 activator or a SIRT1 inhibitor can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985. The composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the subject being treated.
  • In pharmaceutical dosage forms, a SIRT1 activator or a SIRT1 inhibitor (“active agent”) may be administered in the form of its pharmaceutically acceptable salts, or an active agent may be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.
  • For oral preparations, an active agent (a SIRT1 activator or a SIRT1 inhibitor) can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • An active agent can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • An active agent can be utilized in aerosol formulation to be administered via inhalation. An active agent can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.
  • Furthermore, an active agent can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. An active agent can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycol monomethyl ethers, which melt at body temperature, yet are solidified at room temperature.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the subject active agent. Similarly, unit dosage forms for injection or intravenous administration may comprise a subject active agent in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • An active agent can be formulated for administration by injection. Typically, injectable compositions are prepared as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. The preparation may also be emulsified or the active ingredient encapsulated in liposome vehicles.
  • In some embodiments, an active agent is delivered by a continuous delivery system. The term “continuous delivery system” is used interchangeably herein with “controlled delivery system” and encompasses continuous (e.g., controlled) delivery devices (e.g., pumps) in combination with catheters, injection devices, and the like, a wide variety of which are known in the art.
  • Mechanical or electromechanical infusion pumps can also be suitable for use with the present invention. Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852; 5,820,589; 5,643,207; 6,198,966; and the like. In general, delivery of active agent can be accomplished using any of a variety of refillable, pump systems. Pumps provide consistent, controlled release over time. In some embodiments, the active agent is in a liquid formulation in a drug-impermeable reservoir, and is delivered in a continuous fashion to the individual.
  • In one embodiment, the drug delivery system is an at least partially implantable device. The implantable device can be implanted at any suitable implantation site using methods and devices well known in the art. An implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned. Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body. Subcutaneous implantation sites are used in some embodiments because of convenience in implantation and removal of the drug delivery device.
  • Drug release devices suitable for use in the invention may be based on any of a variety of modes of operation. For example, the drug release device can be based upon a diffusive system, a convective system, or an erodible system (e.g., an erosion-based system). For example, the drug release device can be an electrochemical pump, osmotic pump, an electroosmotic pump, a vapor pressure pump, or osmotic bursting matrix, e.g., where the drug (“active agent”) is incorporated into a polymer and the polymer provides for release of drug formulation concomitant with degradation of a drug-impregnated polymeric material (e.g., a biodegradable, drug-impregnated polymeric material). In other embodiments, the drug release device is based upon an electrodiffusion system, an electrolytic pump, an effervescent pump, a piezoelectric pump, a hydrolytic system, etc.
  • Drug release devices based upon a mechanical or electromechanical infusion pump can also be suitable for use with the present invention. Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852, and the like. In general, a subject treatment method can be accomplished using any of a variety of refillable, non-exchangeable pump systems. Pumps and other convective systems are generally preferred due to their generally more consistent, controlled release over time. Osmotic pumps are used in some embodiments due to their combined advantages of more consistent controlled release and relatively small size (see, e.g., PCT published application no. WO 97/27840 and U.S. Pat. Nos. 5,985,305 and 5,728,396)). Exemplary osmotically-driven devices suitable for use in the invention include, but are not necessarily limited to, those described in U.S. Pat. Nos. 3,760,984; 3,845,770; 3,916,899; 3,923,426; 3,987,790; 3,995,631; 3,916,899; 4,016,880; 4,036,228; 4,111,202; 4,111,203; 4,203,440; 4,203,442; 4,210,139; 4,327,725; 4,627,850; 4,865,845; 5,057,318; 5,059,423; 5,112,614; 5,137,727; 5,234,692; 5,234,693; 5,728,396; and the like.
  • In some embodiments, the drug delivery device is an implantable device. The drug delivery device can be implanted at any suitable implantation site using methods and devices well known in the art. As noted infra, an implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned. Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body.
  • In some embodiments, an active agent (a SIRT1 activator or a SIRT1 inhibitor) is delivered using an implantable drug delivery system, e.g., a system that is programmable to provide for administration of an active agent. Exemplary programmable, implantable systems include implantable infusion pumps. Exemplary implantable infusion pumps, or devices useful in connection with such pumps, are described in, for example, U.S. Pat. Nos. 4,350,155; 5,443,450; 5,814,019; 5,976,109; 6,017,328; 6,171,276; 6,241,704; 6,464,687; 6,475,180; and 6,512,954. A further exemplary device that can be adapted for the present invention is the Synchromed infusion pump (Medtronic).
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985. The composition or formulation to be administered will, in any event, contain a quantity of an active agent adequate to achieve the desired state in the subject being treated.
  • Oral Formulations
  • In some embodiments, an active agent (a SIRT1 activator or a SIRT1 inhibitor) is formulated for oral delivery to an individual in need of such an agent.
  • For oral delivery, a formulation comprising an active agent will in some embodiments include an enteric-soluble coating material. Suitable enteric-soluble coating material include hydroxypropyl methylcellulose acetate succinate (HPMCAS), hydroxypropyl methyl cellulose phthalate (HPMCP), cellulose acetate phthalate (CAP), polyvinyl phthalic acetate (PVPA), Eudragit™, and shellac.
  • As one non-limiting example of a suitable oral formulation, an active agent is formulated with one or more pharmaceutical excipients and coated with an enteric coating, as described in U.S. Pat. No. 6,346,269. For example, a solution comprising an active agent and a stabilizer is coated onto a core comprising pharmaceutically acceptable excipients, to form an active agent-coated core; a sub-coating layer is applied to the active agent-coated core, which is then coated with an enteric coating layer. The core generally includes pharmaceutically inactive components such as lactose, a starch, mannitol, sodium carboxymethyl cellulose, sodium starch glycolate, sodium chloride, potassium chloride, pigments, salts of alginic acid, talc, titanium dioxide, stearic acid, stearate, micro-crystalline cellulose, glycerin, polyethylene glycol, triethyl citrate, tributyl citrate, propanyl triacetate, dibasic calcium phosphate, tribasic sodium phosphate, calcium sulfate, cyclodextrin, and castor oil. Suitable solvents for an active agent include aqueous solvents. Suitable stabilizers include alkali-metals and alkaline earth metals, bases of phosphates and organic acid salts and organic amines. The sub-coating layer comprises one or more of an adhesive, a plasticizer, and an anti-tackiness agent. Suitable anti-tackiness agents include talc, stearic acid, stearate, sodium stearyl fumarate, glyceryl behenate, kaolin and aerosil. Suitable adhesives include polyvinyl pyrrolidone (PVP), gelatin, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), vinyl acetate (VA), polyvinyl alcohol (PVA), methyl cellulose (MC), ethyl cellulose (EC), hydroxypropyl methyl cellulose phthalate (HPMCP), cellulose acetate phthalates (CAP), xanthan gum, alginic acid, salts of alginic acid, Eudragit™ copolymer of methyl acrylic acid/methyl methacrylate with polyvinyl acetate phthalate (PVAP). Suitable plasticizers include glycerin, polyethylene glycol, triethyl citrate, tributyl citrate, propanyl triacetate and castor oil. Suitable enteric-soluble coating material include hydroxypropyl methylcellulose acetate succinate (HPMCAS), hydroxypropyl methyl cellulose phthalate (HPMCP), cellulose acetate phthalate (CAP), polyvinyl phthalic acetate (PVPA), Eudragit™ and shellac.
  • Suitable oral formulations also include an active agent formulated with any of the following: microgranules (see, e.g., U.S. Pat. No. 6,458,398); biodegradable macromers (see, e.g., U.S. Pat. No. 6,703,037); biodegradable hydrogels (see, e.g., Graham and McNeill (1989) Biomaterials 5:27-36); biodegradable particulate vectors (see, e.g., U.S. Pat. No. 5,736,371); bioabsorbable lactone polymers (see, e.g., U.S. Pat. No. 5,631,015); slow release protein polymers (see, e.g., U.S. Pat. No. 6,699,504; Pelias Technologies, Inc.); a poly(lactide-co-glycolide/polyethylene glycol block copolymer (see, e.g., U.S. Pat. No. 6,630,155; Atrix Laboratories, Inc.); a composition comprising a biocompatible polymer and particles of metal cation-stabilized agent dispersed within the polymer (see, e.g., U.S. Pat. No. 6,379,701; Alkermes Controlled Therapeutics, Inc.); and microspheres (see, e.g., U.S. Pat. No. 6,303,148; Octoplus, B. V.).
  • Suitable oral formulations also include an active agent with any of the following: a carrier such as Emisphere® (Emisphere Technologies, Inc.); TIMERx, a hydrophilic matrix combining xanthan and locust bean gums which, in the presence of dextrose, form a strong binder gel in water (Penwest); Geminex™ (Penwest); Procise™ (GlaxoSmithKline); SAVIT™ (Mistral Pharma Inc.); RingCap™ (Alza Corp.); Smartrix® (Smartrix Technologies, Inc.); SQZgel™ (MacroMed, Inc.); Geomatrix™ (Skye Pharma, Inc.); Oros® Tri-layer (Alza Corporation); and the like.
  • Also suitable for use are formulations such as those described in U.S. Pat. No. 6,296,842 (Alkermes Controlled Therapeutics, Inc.); U.S. Pat. No. 6,187,330 (Scios, Inc.); and the like.
  • Also suitable for use herein are formulations comprising an intestinal absorption enhancing agent, and an active agent. Suitable intestinal absorption enhancers include, but are not limited to, calcium chelators (e.g., citrate, ethylenediamine tetracetic acid); surfactants (e.g., sodium dodecyl sulfate, bile salts, palmitoylcarnitine, and sodium salts of fatty acids); toxins (e.g., zonula occludens toxin); and the like.
  • Inhalational Formulations
  • An active agent (a SIRT1 activator or a SIRT1 inhibitor) will in some embodiments be administered to a patient by means of a pharmaceutical delivery system for the inhalation route. An active agent can be formulated in a form suitable for administration by inhalation. The inhalational route of administration provides the advantage that the inhaled drug can bypass the blood-brain barrier. The pharmaceutical delivery system is one that is suitable for respiratory therapy by delivery of an active agent to mucosal linings of the bronchi. This invention can utilize a system that depends on the power of a compressed gas to expel the active agent from a container. An aerosol or pressurized package can be employed for this purpose.
  • As used herein, the term “aerosol” is used in its conventional sense as referring to very fine liquid or solid particles carries by a propellant gas under pressure to a site of therapeutic application. When a pharmaceutical aerosol is employed in this invention, the aerosol contains an active agent, which can be dissolved, suspended, or emulsified in a mixture of a fluid carrier and a propellant. The aerosol can be in the form of a solution, suspension, emulsion, powder, or semi-solid preparation. Aerosols employed in the present invention are intended for administration as fine, solid particles or as liquid mists via the respiratory tract of a patient. Various types of propellants known to one of skill in the art can be utilized. Suitable propellants include, but are not limited to, hydrocarbons or other suitable gas. In the case of the pressurized aerosol, the dosage unit may be determined by providing a value to deliver a metered amount.
  • An active agent can also be formulated for delivery with a nebulizer, which is an instrument that generates very fine liquid particles of substantially uniform size in a gas. For example, a liquid containing the active agent is dispersed as droplets. The small droplets can be carried by a current of air through an outlet tube of the nebulizer. The resulting mist penetrates into the respiratory tract of the patient.
  • A powder composition containing an active agent, with or without a lubricant, carrier, or propellant, can be administered to a mammal in need of therapy. This embodiment of the invention can be carried out with a conventional device for administering a powder pharmaceutical composition by inhalation. For example, a powder mixture of the compound and a suitable powder base such as lactose or starch may be presented in unit dosage form in for example capsular or cartridges, e.g. gelatin, or blister packs, from which the powder may be administered with the aid of an inhaler.
  • There are several different types of inhalation methodologies which can be employed in connection with the present invention. An active agent can be formulated in basically three different types of formulations for inhalation. First, an active agent can be formulated with low boiling point propellants. Such formulations are generally administered by conventional meter dose inhalers (MDI's). However, conventional MDI's can be modified so as to increase the ability to obtain repeatable dosing by utilizing technology which measures the inspiratory volume and flow rate of the patient as discussed within U.S. Pat. Nos. 5,404,871 and 5,542,410.
  • Alternatively, an active agent can be formulated in aqueous or ethanolic solutions and delivered by conventional nebulizers. In some embodiments, such solution formulations are aerosolized using devices and systems such as disclosed within U.S. Pat. Nos. 5,497,763; 5,544,646; 5,718,222; and 5,660,166.
  • An active agent can be formulated into dry powder formulations. Such formulations can be administered by simply inhaling the dry powder formulation after creating an aerosol mist of the powder. Technology for carrying such out is described within U.S. Pat. No. 5,775,320 and U.S. Pat. No. 5,740,794.
  • Dosages and Dosing
  • Depending on the subject and condition being treated and on the administration route, an active agent can be administered in dosages of, for example, 0.1 μg to 500 mg/kg body weight per day, e.g., from about 0.1 μg/kg body weight per day to about 1 μg/kg body weight per day, from about 1 μg/kg body weight per day to about 25 μg/kg body weight per day, from about 25 μg/kg body weight per day to about 50 μg/kg body weight per day, from about 50 μg/kg body weight per day to about 100 μg/kg body weight per day, from about 100 μg/kg body weight per day to about 500 μg/kg body weight per day, from about 500 μg/kg body weight per day to about 1 mg/kg body weight per day, from about 1 mg/kg body weight per day to about 25 mg/kg body weight per day, from about 25 mg/kg body weight per day to about 50 mg/kg body weight per day, from about 50 mg/kg body weight per day to about 100 mg/kg body weight per day, from about 100 mg/kg body weight per day to about 250 mg/kg body weight per day, or from about 250 mg/kg body weight per day to about 500 mg/kg body weight per day. The range is broad, since in general the efficacy of a therapeutic effect for different mammals varies widely with doses typically being 20, 30 or even 40 times smaller (per unit body weight) in man than in the rat. Similarly the mode of administration can have a large effect on dosage. Thus, for example, oral dosages may be about ten times the injection dose. Higher doses may be used for localized routes of delivery.
  • For example, an active agent can be administered in an amount of from about 1 mg to about 1000 mg per dose, e.g., from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 50 mg, from about 50 mg to about 75 mg, from about 75 mg to about 100 mg, from about 100 mg to about 125 mg, from about 125 mg to about 150 mg, from about 150 mg to about 175 mg, from about 175 mg to about 200 mg, from about 200 mg to about 225 mg, from about 225 mg to about 250 mg, from about 250 mg to about 300 mg, from about 300 mg to about 350 mg, from about 350 mg to about 400 mg, from about 400 mg to about 450 mg, from about 450 mg to about 500 mg, from about 500 mg to about 750 mg, or from about 750 mg to about 1000 mg per dose.
  • An exemplary dosage may be a solution suitable for intravenous administration; a tablet taken from two to six times daily, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active agent, etc. The time-release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release.
  • Those of skill in the art will readily appreciate that dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means.
  • Although the dosage used will vary depending on the clinical goals to be achieved, a suitable dosage range is in some embodiments one which provides up to about 1 μg to about 1,000 μg or about 10,000 μg of active agent in a blood sample taken from the individual being treated, about 24 hours after administration of the active agent to the individual.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more active agents. Similarly, unit dosage forms for injection or intravenous administration may comprise the active agent(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • In some embodiments, multiple doses of an active agent are administered. The frequency of administration of an active agent can vary depending on any of a variety of factors, e.g., severity of the symptoms, etc. For example, in some embodiments, an active agent is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid). As discussed above, in some embodiments, an active agent is administered continuously.
  • The duration of administration of an active agent, e.g., the period of time over which an active agent is administered, can vary, depending on any of a variety of factors, e.g., patient response, etc. For example, an active agent can be administered over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more. In some embodiments, an active agent is administered for the lifetime of the individual.
  • Routes of Administration
  • An active agent is administered to an individual using any available method and route suitable for drug delivery, including in vivo and ex vivo methods, as well as systemic and localized routes of administration. Administration can be acute (e.g., of short duration, e.g., a single administration, administration for one day to one week), or chronic (e.g., of long duration, e.g., administration for longer than one week, e.g., administration over a period of time of from about 2 weeks to about one month, from about one month to about 3 months, from about 3 months to about 6 months, from about 6 months to about 1 year, or longer than one year).
  • Conventional and pharmaceutically acceptable routes of administration include intranasal, intramuscular, intratracheal, subcutaneous, intradermal, transdermal, sublingual, topical application, intravenous, ocular (e.g., topically to the eye, intravitreal, etc.), rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration may be combined, if desired, or adjusted depending upon the agent and/or the desired effect. The active agent can be administered in a single dose or in multiple doses.
  • An active agent can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes. In general, routes of administration contemplated by the invention include, but are not necessarily limited to, enteral, parenteral, and inhalational routes.
  • Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular, intraorbital, intracapsular, intraspinal, intrasternal, ocular, and intravenous routes, i.e., any route of administration other than through the alimentary canal. Parenteral administration can be carried to effect systemic or local delivery of the agent. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
  • An active agent can also be delivered to the subject by enteral administration. Enteral routes of administration include, but are not necessarily limited to, oral and rectal (e.g., using a suppository) delivery.
  • Methods of administration of an active agent through the skin or mucosa include, but are not necessarily limited to, topical application of a suitable pharmaceutical preparation, transdermal transmission, injection and epidermal administration. For transdermal transmission, absorption promoters or iontophoresis are suitable methods. Iontophoretic transmission may be accomplished using commercially available “patches” which deliver their product continuously via electric pulses through unbroken skin for periods of several days or more.
  • Subjects Suitable for Treatment
  • Subjects suitable for treatment with a subject method include individuals in need of such treatment.
  • Methods of Reducing Chronic Immune Hyperactivity
  • Subjects suitable for treatment with a subject method of reducing chronic immune hyperactivity include individuals who have chronic immune hyperactivity due to an infection with an immunodeficiency virus, e.g., human immunodeficiency virus-1, human immunodeficiency virus-2.
  • Methods of Increasing an Immune Response
  • Subjects suitable for treatment a subject method of increasing an immune response include an individual who has been infected with a pathogenic microorganism; an individual who is susceptible to infection by a pathogenic microorganism, but who has not yet been infected; and an individual who has cancer.
  • Subjects suitable for treatment with a subject method of increasing an immune response include pediatric target populations, e.g., individuals between about 1 year of age and about 17 years of age, including infants (e.g., from about 1 month old to about 1 year old); children (e.g., from about 1 year old to about 12 years old); and adolescents (e.g., from about 13 years old to about 17 years old). Subjects suitable for treatment with a subject method of increasing an immune response include adult individuals.
  • Subjects suitable for treatment with a subject method of increasing an immune response include immunodeficient individuals, e.g., individuals with an acquired immunodeficiency, e.g., as a results of radiation therapy for cancer, as a result of corticosteroid treatment, as a result of cancer chemotherapy, etc. Also suitable for treatment with a subject method of increasing an immune response are individuals who have undergone bone marrow transplantation or any other organ transplantation, e.g., immunosuppressed individuals.
  • EXAMPLES
  • The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pl, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal(ly); s.c., subcutaneous(ly); and the like. “a” preceding a protein denotes an antibody to that protein; for example, α-p65 is an antibody to p65.
  • Example 1 Human Immunodeficiency Virus Type 1 Tat Protein Inhibits the SIRT1 Deacetylase and Induces T Cell Hyperactivation Experimental Procedures Cells and Plasmids
  • HeLa, 293, 293T and Jurkat cells were cultured under standard tissue culture conditions. MEF cells derived from SIRT1−/− cells were grown as described (Pagans et al., 2005). Mutant constructs for SIRT1 HDAC domain were prepared by site-directed mutagenesis using FLAG-tagged SIRT1 as a template. The Sir2α deletion constructs were kindly provided by V. Sartorelli, NIH, the 3×-κB luciferase reporter by N. Chirmule, Merck Inc, the IκBa luciferase reporter by K. Yamamoto, UCSF, and the E-selectin luciferase reporter by J. Pober, Yale University.
  • Infection with Lentiviral Vectors
  • LTR-GFP and LTR-Tat-GFP are HIV-based vectors derived from the pHR′ series, in which GFP alone or Tat (101 aa) and GFP are under the control of the HIV-1 LTR through the use of an internal ribosomal entry site (Jordan et al., 2001). For experiments with mutant Tat, the EF-1α promoter was inserted into the LTR-GFP vector upstream of wild type or mutant Tat (EF1α-Tat-GFP). Each vector was cotransfected into 293T cells together with a packaging construct (pCMV-R8.91) that provides all HIV genes required for production of infective particles and a plasmid encoding the vesicular stomatitis virus envelope G protein (VSV-G) to produce pseudotyped viral particles with broad host range and high infectivity. Viral supernatant containing 1500 ng of p24 was used to infect 18×106 Jurkat T cells. Infections were carried out in 6-well plates at 2400 rpm in a Beckman-Coulter centrifuge for 2 h at 32° C. in the presence of polybrene (1 μg/ml, Sigma). Jurkat T cells (106) were stimulated with plate bound α-CD3 (3 μg/ml) and soluble α-CD28 (1 μg/ml) antibodies 36 h after infection as previously described (Ott et al., 1998) or were preincubated with nicotinamide (10 mM, Sigma) for 1 h before treatment with α-CD3/28 antibodies. Flow cytometry analysis of GFP expression (FACSCalibur, BD Bioscience) showed that infection efficiencies ranged from 70-98% GFP cells in individual experiments. Tat expression was visualized by western blotting with polyclonal α-FLAG and α-β-actin (Sigma) antibodies.
  • Generation of SIRT1- and Tat-Expressing MEF Cell Lines
  • Open reading frames corresponding to Myc-tagged human SIRT1 and T7-tagged HIV-1 Tat (101 aa) were inserted into the murine stem cell virus (MSCV)-based retroviral vectors MSCV-puromycin and MSCV-zeocin, respectively (Clontech). To obtain recombinant virus, 10 μg of each construct or empty control vectors were transfected into BOSC23 cells, a retroviral packaging cell line derived from 293 cells. The supernatants were collected 48 h after transfection and filtered through a 0.45-μm membrane. SIRT1−/− MEF cells (2×105) were incubated with 2 ml of the supernatant containing SIRT1-expressing retrovirus or control virus together with polybrene (8 μg/ml, Sigma). Cells were selected after 48 h with puromycin (2.5 μg/ml, Invitrogen). Polyclonal puromycin-resistant MEF cells were reinfected with Tat-expressing or empty control retroviruses and selected in the presence of both puromycin (2.5 μg/ml) and zeocin (100 μg/ml, Invitrogen).
  • RNA Purification, cDNA Synthesis, and Real-Time RT-PCR
  • Total RNA was extracted using RNA STAT-60 reagent (Tel-Test) according to the manufacturer's instruction. The first strand cDNA was generated using 2 μg of total RNA and SuperScript reverse transcriptase (Invitrogen). Human IL-2 and murine E-selectin mRNAs were quantified by QuantiTect gene expression assays (Qiagen). Murine IκBα and murine/human GAPDH mRNA concentrations were determined with SYBR Green I master mix (MCLab). Relative gene expression ratios between stimulated and nonstimulated conditions were calculated by the equation, ratio=2−ΔΔCt, comparative Ct method Applied Biosystems).
  • Transfections and Coimmunoprecipitations
  • Luciferase reporter constructs and protein expression vectors were transiently cotransfected into HeLa cells using Lipofectamine (Invitrogen). 24 h after transfection, cells were lysed and processed for luciferase assays (Promega). P values (paired t-test) were used for statistical analysis.
  • 293 cells grown in 6-well plates at 70% confluency were cotransfected with expression vectors indicated in the Figure legends. Transfection, cell lysis, immunoprecipitation, and western blotting were previously described (Pagans et al., 2005).
  • Fluorescent Deacetylase Assay
  • Full-length human SIRT1 (His-tagged) was purified using Ni-NTA agarose (Qiagen) and Superdex 200 gel filtration. The rate of 1 μM SIRT1 was calculated for 200 μM fluorogenic deacetylase substrate (BioMol) in the presence of twelve concentrations (1, 2, 3, 4, 5, 6, 10, 15, 25, 50, 126, 252 μM) of synthetic Tat (72 aa; Peptide Specialty Laboratory, Heidelberg, Germany).
  • Radioactive HDAC Assays
  • Expression plasmids for FLAG-tagged wild type and mutants SIRT1 (1 μg) were transfected in 293 cells with lipofectamine reagent. Equal amounts of whole cell extracts (2 mg) were immunoprecipitated and in vitro HDAC assays were performed as previously described (Pagans et al., 2005).
  • Recombinant p65 protein prepared from baculovirus-infected Sf9 insect cells (BD Biosciences) was acetylated by immunoprecipitated p300 overexpressed in 293 cells as previously described (Chen et al., 2005). Acetylated p65 protein was incubated with recombinant SIRT1 (1 μg/5 U; Biomol) in SIRT1 deacetylase buffer (50 mM Tris- HCl pH 9, 4 mM MgCl2, 0.2 mM DTT) in the presence of NAD (1 mM; Sigma) for 3 h at 37° C. Reactions containing synthetic Tat (0.1 μg) or nicotinamide (10 mM) were preincubated for 15 mM at room temperature. Reactions were stopped by the addition of SDS loading buffer, boiled, and after brief centrifugation, analyzed by western blotting with rabbit α-AcK310 p65 (Chen et al., 2005) or mouse α-p65 antibodies (sc-8008, Santa Cruz).
  • Results Tat and Nicotinamide Hyperactivate T Cells Via the Same Cellular Pathway
  • To recapitulate the effect of Tat on immune activation in a manner as close to natural HIV infection as possible, we employed an HIV-based lentiviral vector in which both FLAG-tagged Tat and green fluorescent protein (GFP) are expressed under the control of the HIV promoter in the 5′-LTR. Jurkat T cells were infected with viral particles containing this vector or a control vector expressing only GFP. Infected cultures were stimulated with antibodies specific for the CD3 and CD28 receptors to mimic physiological T-cell activation.
  • Tat expression was visualized by western blot analysis (FIG. 1A). Treatment with α-CD3/28 slightly increased the levels of Tat due to the stimulatory effect of these antibodies on the viral LTR. In cells expressing Tat, a ˜1200-fold induction of IL-2 mRNA was detected by real-time PCR in response to α-CD3/28 treatment, whereas IL-2 expression in control cells was only induced ˜250 fold (FIG. 1B). A similar difference was observed when IL-2 protein was measured in the culture supernatant (FIG. 1C). No effect of Tat was observed in nonactivated cultures. These findings recapitulated results previously obtained in peripheral blood lymphocytes and Jurkat T cells infected with infectious HIV and support a model where Tat synergistically stimulates IL-2 production following ligation of the CD3 and CD28 receptors (Fortin et al., 2004; Ott et al., 1997).
  • To test the potential regulatory role of SIRT1 in the IL-2 response, we treated Jurkat T cells with nicotinamide, a natural byproduct of the sirtuin deacetylase reaction which functions as a feedback inhibitor of these enzymes (Landry et al., 2000). Treatment with nicotinamide superinduced IL-2 mRNA to similar levels as Tat in α-CD3/28-stimulated cells (FIG. 1D). To test whether Tat and nicotinamide synergize in the hyperactivation of IL-2 expression, we treated Tat- or control-infected Jurkat cultures with nicotinamide before activation. These studies revealed no additive effect of combinations of Tat and nicotinamide on IL-2 expression in α-CD3/28-stimulated T cells (FIG. 1E). Levels of Tat protein were equivalent in infected cultures treated with nicotinamide and control-treated cultures excluding the possibility that nicotinamide suppressed the expression of Tat (FIG. 1F). These results suggest that Tat and nicotinamide may target closely related cellular pathways and raised the interesting possibility that Tat hyperactivation of T cells may involve its known assembly with SIRT1.
  • FIGS. 1A-F. Tat and nicotinamide cause T-cell hyperactivation (A) Western blot analysis of Tat protein in Jurkat T cells infected with lentiviral vectors expressing LTR-GFP and LTR-Tat-GFP. Infected cultures were activated for 2 h and 8 h with α-CD3/28 antibodies. (B) Real-time RT-PCR analysis of IL-2 mRNA levels in infected cultures 2 h after activation with α-CD3/28. Results are expressed as fold induction by α-CD3/28 treatment. (C) IL-2 protein levels measured by ELISA in the supernatant of infected cultures 8 h after α-CD3/28 treatment. (D) Real-time RT-PCR analysis of IL-2 mRNA levels in noninfected Jurkat T cells after treatment with nicotinamide and activation with α-CD3/28. (E) Real-time RT-PCR analysis of IL-2 mRNA levels in Jurkat T cells infected with lentiviral vectors after treatment with nicotinamide and activation with α-CD3/28 antibodies. (F) Western blot analysis of Tat protein in Jurkat T cells infected with lentiviral vectors after treatment with nicotinamide and activation with α-CD3/28. In B-E averages of three independent experiments (±SEM) are shown.
  • SIRT1 is Required for the Hyperactivation of NF-κB Target Genes by Tat
  • T-cell activation is coupled to the stimulation of several transcription factors that regulate the transcriptional status of the IL-2 gene. Efforts were focused on NF-κB since its activity is regulated by SIRT1 (Yeung et al., 2004). It was previously previously reported that Tat enhanced IL-2 promoter activity via the NF-κB binding sites present within the IL-2 promoter (Fortin et al., 2004; Ott et al., 1997; Ott et al., 1998; Westendorp et al., 1994). It was speculated that Tat, like nicotinamide, inhibits the SIRT1 deacetylase activity leading to hyperactivation of NF-κB-responsive genes including IL-2.
  • This hypothesis was tested in cells derived from SIRT1−/− mice. Mouse embryonic fibroblasts (MEFs) lacking SIRT1 expression were infected with a retroviral vector expressing SIRT1 or the empty vector alone. Polyclonal populations of SIRT1-expressing or SIRT1-negative cells were each infected with retroviral vectors expressing Tat or with the empty vector alone. This protocol generated four polyclonal MEF cultures, allowing the analysis of the effect of Tat in the presence or absence of SIRT1 (FIG. 2A). MEF cell lines were treated with TNFα to induce NF-κB. Since MEF cells do not produce IL-2, we focused instead on the expression of IκBα, a ubiquitous NF-κB-responsive gene product (Sun et al., 1993). We found that Tat expression increased endogenous levels of IκBα mRNA 2 to 4-fold over control-infected cells in response to TNFα (FIG. 2B). Remarkably, this effect was only observed in cultures where SIRT1 expression had been reconstituted. No difference between Tat-expressing and control cells was detected in SIRT1-negative cells, indicating that Tat-mediated superinduction of NF-κB-responsive genes was dependent on SIRT1.
  • A similar result was observed with E-selectin, another NF-κB-responsive gene that is also superinduced by Tat (Cota-Gomez et al., 2002; Lee et al., 2004). After TNFα treatment, endogenous E-selectin gene expression was enhanced 6-fold by Tat in SIRT1-expressing, but not in SIRT1-negative cells (FIG. 2C). Here, the Tat effect was only observed at 2 h after TNFα treatment. At 8 h, no induction of E-selectin gene expression was evident, consistent with the prior finding that E-selectin mRNA accumulation peaks 2 h after TNFα treatment and then rapidly declines (Edelstein et al., 2005). In contrast to the results with IκBα and E-selectin, Tat had no effect on the constitutive expression of the endogenous glyceraldehyde-3-phosphate dehydrogenase gene (GAPDH) (FIG. 2D).
  • FIGS. 2A-D. Tat-mediated superinduction of NF-κB-responsive genes requires SIRT1 (A) Western blot analysis of SIRT1, Tat and β-actin in SIRT1−/− MEF cells infected with MSCV-based retroviral particles expressing SIRT1 or Tat. (B-D) Real-time RT-PCR analysis of IκBα (B), E-selectin (C), and GAPDH (D) mRNA levels in the four polyclonal MEF populations described in (A) after incubation with TNFα (20 ng/ml) for 2 h and 8 h. Data are presented relative to values obtained in cells lacking Tat (100%). The mean of three independent experiments (±SEM) is shown.
  • Tat Neutralizes the Negative Effect of SIRT1 on NF-κB Function
  • Next, we assessed the effect of Tat on the IκBα and E-selectin gene promoters. We transfected an IκBα promoter luciferase reporter construct together with combinations of expression vectors encoding p65, SIRT1, and Tat into HeLa cells (FIG. 3A). SIRT1 suppressed the activation of the IκBα promoter by p65 in accordance with the previous finding that deacetylation by SIRT1 inactivates p65 activity (Yeung et al., 2004). Coexpression of Tat restored p65 activity, suggesting that Tat interfered with the deacetylation of p65 by SIRT1 (FIG. 3A). No change in expression levels of p65 was observed by western blotting in the presence of SIRT1 or Tat. The suppressive activity of SIRT1 was dependent on its intrinsic deacetylase activity of SIRT1, since a catalytically inactive SIRT1 mutant (P447E; shown in FIG. 4D) did not suppress the activity of p65 and was unresponsive to the action of Tat. Expression of Tat did not affect basal or p65-mediated activity of the IκBα promoter in the absence of SIRT1 expression (FIG. 3A).
  • The same results were observed using an E-selectin promoter luciferase construct. Again, SIRT1 reduced p65-activated E-selectin promoter activity, and Tat reversed the negative effect of SIRT1 (FIG. 3B). No reduction was observed with the mutant SIRT1, which was also unresponsive to Tat (FIG. 3B).
  • To verify that Tat and SIRT1 modulated the activities of the E-selectin and IκBα promoters through NF-κB, we transfected p65-, Tat-, and SIRT1-expressing constructs together with a 3×-κB luciferase reporter. Expression of SIRT1 reduced p65-mediated activation of the κB reporter, while coexpression of Tat reversed this negative effect (FIG. 3C). Again, expression of the catalytically inactive SIRT1 mutant did not interfere with the activity of p65, and additional Tat expression had no effect in this setting (FIG. 3C). Likewise, overexpression of a p65 mutant (K310R), which lacked the deacetylation site for SIRT1 and showed reduced activation of the NF-κB luciferase construct, was unaffected by SIRT1 and Tat (FIG. 3C). These results demonstrate that Tat targets the SIRT1 deacetylase activity to hyperactivate NF-κB function.
  • FIGS. 3A-C. Tat neutralizes the inhibitory effect of SIRT1 on NF-κB-responsive promoters. Promoter reporter assays using (A) the IκBα, (B) the E-selectin, and (C) the 3×-κB luciferase reporter construct. Luciferase constructs (0.2 μg) were transiently cotransfected with expression vectors for p65 or p65 K310R (1 ng), SIRT1 or SIRT1 P447E (0.2 μg), and Tat (50 ng) into HeLa cells. The mean of three independent experiments (±SEM) is shown.
  • Tat Binds the Deacetylase Domain of SIRT1
  • We showed previously that Tat and SIRT1 interact in cells (Pagans et al., 2005). To map the Tat-interacting domain in SIRT1, we used deletion mutants of the murine SIRT1 protein (Sir2α) (Fulco et al., 2003). Progressive N-terminal deletions at amino acids 120, 236, or 341 did not affect coimmunoprecipitation of SIRT1 with Tat (FIG. 4A, lanes 1-3). However, deletion of the first 512 amino acids abrogated binding to Tat, indicating that the Tat-interaction domain lies between amino acids 341 and 512 of SIRT1 (FIG. 4A, lane 4). In agreement with this conclusion, expression of amino acids 236 to 510 of SIRT1 proved sufficient to support interaction with Tat (FIG. 4A, lane 5). The results of the coimmunoprecipitation studies are summarized in FIG. 4B.
  • The region spanning amino acids 341-512 contains the catalytic domain of SIRT1, which is shared among all sirtuins. It is 98% conserved in the murine and human SIRT1 proteins and consists of two distinct domains: a large Rossmann fold domain characteristic of NAD+-binding proteins and a smaller domain containing a structural zinc ion. The NAD and acetyl-lysine substrate bind in the active site cleft between the two domains (Avalos et al., 2002). To determine whether Tat interacts with the catalytic domain of SIRT1, point mutations were introduced into human SIRT1 expression construct to disrupt NAD+-binding (R274A, N346A) or acetyl-lysine binding (F414D, E416A, V445E, P447E) (FIG. 4C). In addition, we mutated two conserved cysteines (C371/374G) located in a zinc-bound domain predicted to serve as a protein-protein interaction domain (Min et al., 2001).
  • Prior to analyzing their interaction with Tat, the catalytic activities of these mutants were measured using an in vitro HDAC assay and radioactive acetylated histone peptides as substrates. We transfected wildtype and mutant SIRT1 proteins into 293 cells and immunoprecipitated the FLAG-tagged proteins (FIG. 4D). The immunoprecipitated material was tested for enzymatic activity. Incubation with wildtype SIRT1 or the R274A mutant resulted in deacetylation of the histone peptide in the presence of NAD (FIG. 4D). The deacetylase activity of the F414D, E416A, and V445E mutants was severely impaired, while no activity was measured for N346A, C371/374G, and P447E mutants.
  • Next, we cotransfected wildtype and SIRT1 mutants together with T7-tagged Tat. Wildtype and mutant SIRT1 proteins were expressed at similar levels (FIG. 4E, upper panel). After pulldown with T7-agarose, immunoprecipitated complexes were analyzed for the presence of SIRT1 (FIG. 4E, lower panels). While binding to Tat was preserved in SIRT1 R274A, N346A, C371/374G, and E416A, no binding to Tat was observed with any of the SIRT1 mutants carrying a mutation within the acetyl-lysine-binding domain (F414D, V445E, P447E). The same results were obtained when SIRT1 proteins were immunoprecipitated, and coimmunoprecipitation of Tat was determined. These data demonstrate that Tat interacts with the substrate-binding domain of SIRT1.
  • FIGS. 4A-E. Tat binds to the acetyl lysine-binding site in SIRT1 (A) Communoprecipitation assay of Tat and SIRT1 deletion mutants. Expression vectors for Myc-tagged murine Sir2α deletion mutants or full length human SIRT1 (each 1 μg) were transiently cotransfected with constructs expressing FLAG-tagged Tat (1 μg) in 293 cells. Immunoprecipitations were performed with α-FLAG agarose and western blotting with α-Myc and α-FLAG antibodies. (B) Schematic summary of Tat binding to murine Sir2α deletion mutants. The relative localization of the human SIRT1 HDAC domain is shown at the bottom. (C) Schematic representation of the human SIRT1 HDAC domain with point mutations. (D) In vitro HDAC assays of immunoprecipitated human SIRT1 mutants with and without NAD+. (E) Communoprecipitation of SIRT1 mutants and Tat in 293 cells. Cell extracts were immunoprecipitated with α-T7 agarose followed by western blotting with α-FLAG and α-T7 antibodies.
  • Tat Inhibits the SIRT1 Deacetylase Activity and Induces Hyperacetylation of NF-κB
  • Since an intact acetyl-lysine binding function is critical for the deacetylase activity of SIRT1, we tested whether Tat binding affected the enzymatic activity of SIRT1. We performed fluorescent deacetylation assays using purified full-length SIRT1 and acetylated p53 peptides (aa 379-382). Addition of synthetic Tat (aa 1-72) inhibited deacetylation of p53 by SIRT1 in a dose-dependent manner (FIG. 5A). The Tat concentration required for half maximal inhibition (IC50) of SIRT1 was 3.5±0.5 μM. In contrast, the IC50 for nicotinamide in the same experiments was 208±21 μM (data not shown). This result indicates that Tat is a very potent inhibitor of the SIRT1 deacetylase activity.
  • Next, we examined the effect of Tat on deacetylation of p65. In vitro acetylated recombinant p65 protein was incubated with recombinant SIRT1 and synthetic Tat proteins. The extent of deacetylation was determined by western blotting with antibody specific for acetylated K310 in p65 (α-AcK310 p65). SIRT1 deacetylated p65 in the absence of Tat (FIG. 5B, lane 2). In the presence of Tat, deacetylation of p65 by SIRT1 was suppressed, confirming that direct interaction between Tat and SIRT1 inhibits the deacetylase activity of SIRT1 (FIG. 5B, lane 3). The same inhibition of p65 deacetylation by SIRT1 was observed with nicotinamide (FIG. 5B, lane 4).
  • To test the effect of Tat on p65 acetylation in cells, we transfected expression vectors for p65, p300, SIRT1 and Tat into 293 cells. Overexpressed p65 translocates spontaneously into the cell nucleus because of limiting amounts of cellular IκBα. To induce efficient p65 acetylation, we coexpressed the acetyltransferase p300 (FIG. 5C, lane 2). Coexpression of SIRT1 reduced acetylation of p65 at K310 caused by p300 as expected (FIG. 5C, lane 3). Introduction of Tat restored p65 acetylation in a dose-response manner (FIG. 5C, lane 4-6). These results demonstrate that Tat reverts deacetylation of p65 by SIRT1 in cells and induces relative hyperacetylation of p65 in the presence of SIRT1.
  • We also examined the acetylation status of endogenous p65 in Tat-expressing cells. 293 cells were tranfected with the Tat expression vector and treated with TNFα to induce nuclear translocation of endogenous p65. Acetylation of endogenous p65 was slightly increased in the presence of Tat (FIG. 5D, lanes 1 and 2). Coexpression of p300 with Tat strongly induced hyperacetylation of p65, consistent with the model that Tat inhibits physiological deacetylation of NF-κB by SIRT1 (FIG. 5D, lanes 3 and 4).
  • FIGS. 5A-D. Tat inhibits the SIRT1 deacetylase activity (A) In vitro deacetylation assay of recombinant SIRT1 in the presence of synthetic Tat (aa 1-72). The rate of 1 μM SIRT1 was calculated for 200 μM fluorogenic deacetylase substrate (aa 379-382 of p53) in the presence of twelve concentrations of synthetic Tat. The data were plotted in Prism with X equal to Log10[Tat] and Y equal to rate. A nonlinear regression was done using the one-site competitive binding equation to determine the IC50. (B) In vitro deacetylase assay of recombinant SIRT1 and recombinant acetylated p65 in the presence of synthetic Tat or nicotinamide. Western blotting was performed with α-AcK310 p65 and α-p65 antibodies. (C) In vivo acetylation assay of overexpressed p65. Expression vectors encoding T7-tagged p65 (0.5 μg), p300 (2 μg), SIRT1 (1 μg), and Tat (0.1, 0.25, and 0.5 μg) were transiently cotransfected in 293 cells as indicated. Total cell lysates were analyzed by western blotting with α-AcK310 p65 or α-T7 antibodies. (D) In vivo acetylation assay of endogenous p65. 293 cells were cotransfected with expression vectors encoding Tat (0.5 μg) and p300 (10 μg) as indicated and treated with TNFα (20 ng/ml) and trichostatin (TSA; 400 nM) over night. TSA is an inhibitor of class I and II HDACs, but not sirtuins and was added to prolong nuclear retention of p65 through hyperacetylation of K218 and K220. Endogenous p65 was immunoprecipitated using α-p65 antibody and subjected to western blotting with α-AcK310 p65 or α-p65 antibodies.
  • Tat Induces T-Cell Hyperactivation Through Hyperacetylation of NF-κB
  • To examine whether the functions of Tat in SIRT1 inhibition and T-cell hyperactivation are linked, we searched for Tat mutants deficient in the induction of p65 hyperacetylation. We screened a series of Tat proteins carrying point mutations in different protein domains of Tat. These point mutants were expressed in 293 cells together with p65 and the p300 acetyltransferase. Expression of p300 induced acetylation of K310 in p65, and wild type Tat (two-exon, 101 aa) proteins further enhanced this acetylation (FIG. 6A, lanes 1-3). Mutations within the so-called cysteine-rich and core domains of Tat (aa 22-31 and aa 38-48) markedly decreased hyperacetylation of p65 induced by Tat, while a point mutation introduced into the N-terminal acidic domain (aa 1-21) still hyperacetylated p65 albeit to a lesser extent (FIG. 6A, lanes 4-7). A Tat mutant containing two point mutations within the RNA-binding domain (aa 49-59) induced p65 hyperacetylation as efficiently as the wild type protein (FIG. 6A, lane 8). Western blotting with FLAG antibody showed that wild type and H13L Tat were equally expressed while the other Tat mutants were slightly less expressed (FIG. 6A, lower panel). Of note, expression levels of K41A and K50/51A Tat were equivalent, and these mutants were chosen for further analysis.
  • To study the effect of Tat mutations on T-cell activation, we generated lentiviral vectors expressing wild type or mutant Tat proteins (K41A and K50/51 A Tat). Because both Tat mutants have reduced transcriptional activities on the HIV LTR, we chose a lentiviral construct, which contained the heterologous elongation factor 1α (EF-1α) promoter driving Tat and GFP expression instead of the HIV LTR. High titer viral stocks were produced and used to infect Jurkat T cells. Flow cytometry of GFP showed equivalent infection with wild type or mutant Tat-expressing viruses (FIG. 6B). Following treatment with α-CD3/28 antibodies, IL-2 mRNA levels were superinduced 2-3 fold by wild type Tat (FIG. 6C). No superinduction was observed with K41A Tat while K50/51A Tat superinduced IL-2 mRNA levels as efficiently as wild type Tat (FIG. 6C). These results collectively demonstrate that Tat expressed in the context of lentiviral infection hyperactivates T cells through hyperacetylation of p65.
  • FIGS. 6A-C. Hyperacetylation of p65 by Tat contributes to T-cell hyperactivation (A) Induction of p65 hyperacetylation by wild type and mutant Tat. Expression vectors encoding T7-tagged p65 (0.5 μg), p300 (2 μg), wild type and mutant Tat (1 μg each) were transiently cotransfected in 293 cells as indicated. Immunoprecipitations were performed with α-T7 agarose or α-FLAG agarose to isolate p65 or Tat followed by western blotting with α-AcK310 p65, α-T7, and α-FLAG antibodies. (B) Flow cytometry of Jurkat T cells infected with lentiviral vectors expressing wild type or mutant Tat and GFP. Data are representative of three independent experiments in which infection efficiencies ranged from 75-85% GFP cells. (C) Real-time RT-PCR analysis of IL-2 mRNA levels induced in infected Jurkat T cells after activation with α-CD3/28 antibodies. Data are presented as fold induction by α-CD3/28 treatment relative to vector (EF-1α-GFP)-infected cells (100%). The average (mean±SEM) of three independent experiments is shown.
  • The data presented above show that Tat binds to the SIRT1 catalytic site and impairs the ability of SIRT1 to deacetylate K310 in p65. It was also found that NF-κB activity is “superactivated” in the presence of Tat. Based on these results, a model is proposed where Tat hyperactivates T cells through inhibition of SIRT1 and abnormally sustained action of NF-κB. In the absence of Tat, a balance of p300 and SIRT1 activities regulates p65 acetylation. This balance tightly controls the activation of cellular genes involved in the immune response of T cells (FIG. 7A). During HIV infection, Tat expression disrupts the balance between p300 and SIRT1 and increases levels of acetylated p65 through inhibition of SIRT1 (FIG. 7B). This increase hyperactivates the function of NF-κB and the expression of NF-κB-responsive genes including IL-2.
  • One of the most studied NF-κB-responsive genes is HIV itself. The two NF-κB-binding sites in the HIV enhancer link HIV transcription to the activation status of the infected T cells. This is particularly important at the beginning of the infectious process and during reactivation from latency when Tat levels are limiting. During these times, the action of cellular transcription factors, most importantly NF-κB, is essential for the production of full-length viral transcripts necessary for the initial synthesis of Tat. Once Tat accumulates to sufficient levels, it binds to TAR RNA and dramatically increases the elongation competence of the RNA polymerase II complex through recruitment of the P-TEFb complex. At that time, Tat may also enhance NF-κB function through inhibition of SIRT1 (FIG. 7B).
  • FIGS. 7A and 7B. Tat blocks the SIRT1 deacetylase activity and superinduces T-cell activation and HIV transcription via NF-κB.
  • Example 2 SIRT1 Activators Suppress Tat-Induced T Cell Hyperactivation
  • Lentiviral vectors, which contained the heterologous elongation factor 1α (EF-1α) promoter driving Tat and GFP expression, were generated. High titer viral stocks were produced and used to infect Jurkat T cells. Flow cytometry of GFP showed equivalent infection with EF-1α-GFP or EF-1α-Tat-GFP viruses. Infected cultures were stimulated with antibodies specific for the CD3 receptor and treated with increased concentrations of resveratrol (0.4 μM, 2 μM, and 10 μM), a known SIRT1 activator for 16 hrs. IL-2 protein was measured in the culture supernatant by ELISA assay. Following stimulation with α-CD3 antibody, IL-2 protein was superinduced ˜4-fold by wild type Tat (FIG. 8A). In cells expressing Tat, ˜2300 pg/ml of IL-2 protein was detected in response to α-CD3 treatment, whereas in control cells, only ˜600 pg/ml of IL-2 protein was induced. Superinduction of IL-2 in Tat-expressing cells was abolished by resveratrol in a concentration dependent manner (FIG. 8A). Treatment of infected cells with 10 μM of resveratrol completely impaired the ability of Tat to superinduce IL-2 production.
  • Resveratrol, a polyphenolic compound present in red wine, functions as a (weak) reversible activator of SIRT1 enzymatic activity (Howitz et al., (2003) Nature 425:191-196., Baur et al., (2006) Nat. Rev. Drug. Discov.). Recently, novel 1000-fold more potent and structurally unrelated small molecule activators of SIRT1 have been developed and have shown potent activity in obesity and insulin resistance (Milne et al., (2007) Nature 450:712-716).
  • The effects of small molecule of SIRT1 (SRT2180) on T cell hyperactivation in Tat-expressing cells were examined. Jurkat T cells were infected with EF-1α-GFP or EF-1α-Tat-GFP viruses (FIG. 8B). Infected cultures were stimulated with α-CD3 antibody and treated with SRT2183 (0.2, 1, and 5 μM) for 16 hrs. Low concentrations of SRT2183 (0.2 and 1 μM) caused increases in IL-2 protein both in control and Tat expressing cells (FIG. 8B). However, higher concentration of SRT2183 (5 μM) suppressed the production of IL-2 protein in Tat expressing cells while slight increase in IL-2 protein was observed in control cells. These results indicate that activation of SIRT1 by either resveratrol or small molecule counterbalance the block in SIRT1 function by Tat and can suppress elevated levels of IL-2 in Tat-expressing cells.
  • FIGS. 8A and 8B. Resveratrol and small molecule activator of SIRT1 (SRT2183) suppresses T cell hyperactivation by Tat. (A) IL-2 protein levels measured by ELISA in the supernatant of infected cultures. 24 hrs after infection with EF1α-GFP and EF1α-Tat-GFP viruses, Jurkat T cells were stimulated with α-CD3 antibody in the presence of increased concentrations of resveratrol (0.4, 2, and 10 μM) for 16 hrs. (B) 24 hrs after infection with EF1α-GFP and EF1α-Tat-GFP viruses, Jurkat T cells were stimulated with α-CD3 antibody in the presence of increased concentrations of SRT2183 (0.2, 1, and 5 μM) for 16 hrs. IL-2 protein levels were measured by ELISA in the supernatant of infected cultures.
  • Example 3 SIRT1 Regulates FoxP3 Stability and Treg Differentiation Through Deacetylation of Three Novel Acetylation Sites in FoxP3
  • The FoxP3 transcription factor is the master regulator of regulatory T cell (Treg) differentiation. High-level FoxP3 expression is required to support the immune suppressive activity of Tregs while Tregs expressing low levels of FoxP3 may cause autoimmunity. Here we show that FoxP3 protein expression is regulated at the posttranscriptional level by the NAD+-dependent SIRT1 deacetylase. Induced Treg differentiation is suppressed when SIRT1 is overexpressed. In contrast, SIRT1 knockdown or treatment with a small molecule inhibitor of SIRT1 (Ex-527) stabilizes FoxP3 protein expression. Importantly, treatment with Ex-527 prevents loss of FoxP3 expression in thymus-derived natural Tregs during ex vivo expansion. Using mass spectrometry of immunoprecipitated FoxP3, we identified three novel acetylation sites in FoxP3 (K31, K262, K267). Mutant FoxP3 proteins, in which the three sites were substituted by arginines, accumulate in cells to higher levels than wildtype FoxP3 and are not further stabilized by treatment with Ex-527. Collectively, our data support a model where SIRT1 deacetylates and destabilizes FoxP3 in Tregs. SIRT1 inhibitors may serve to optimize Treg-based approaches to treat autoimmune diseases or graft rejections.
  • Results
  • SIRT1 Plays a Negative Role in the Differentiation of Induced Tregs (iTregs)
  • To gain insight into the role of SIRT1 in Treg biology, SIRT1 protein expression during ex vivo Treg differentiation was analyzed. Naïve CD4+ T cells (CD4+CD25−CD69−CD44−) were isolated from mouse spleen and lymph nodes and were activated by anti-CD3/CD28 antibodies alone or in combination with cytokines driving differentiation into T helper (Th) 1, Th2 or induced Treg (iTreg) cells. While SIRT1 expression was increased upon CD3/CD28 activation, it was decreased after T cell differentiation, most noticeably in iTregs (FIG. 9A, lane 5).
  • To test whether SIRT1 plays a role in iTreg differentiation, SIRT1 was overexpressed during iTreg differentiation. Activated CD4+ cells were infected with bicistronic retroviral vectors expressing SIRT1 and the Thy1.1 surface marker (MSCV-SIRT1-Thy1.1) or a control vector expressing Thy1.1 alone (MSCV-Thy1.1). Cell were further differentiated into Tregs in the presence of recombinant human TGF-β and recombinant human IL-2 as previously described (Fu et al., 2004). After 3 days, cells were stained for surface Thy1.1 and intracellular FoxP3 and analyzed by flow cytometry. No difference in the number of FoxP3+Thy1.1+ cells was observed in cultures expressing SIRT1 or control cells; however, the mean fluorescence intensity (MFI) of FoxP3 was decreased in cells overexpressing SIRT1 (FIG. 9B). Cells were cultured 3 more days in IL-2 alone and were reanalyzed at day 6 after infection. Both, the number of FoxP3+/Thy1.1+ cells and FoxP3 MFI were decreased in SIRT1-expressing cells (FIG. 9B). The opposite result was observed when SIRT1 was knocked down during iTreg differentiation using lentiviral shRNA vectors. Collectively, these data indicate that SIRT1 decreases the expression of FoxP3 protein and negatively regulates iTreg differentiation.
  • Binding and Deacetylation of FoxP3 by SIRT1
  • To test whether SIRT1 controls FoxP3 expression through deacetylation coimmunoprecipitation experiments were performed in 293 T cells. Hemagglutinin (HA)-tagged SIRT1 protein coimmunoprecipitated with FLAG-tagged FoxP3 in cells transfected with SIRT1- and FoxP3 expression vectors, but no signal was obtained when SIRT1 or FoxP3 were expressed alone (FIG. 10A). Next, a series of deletion mutants of FoxP3 (Ono et al., 2007) was tested, to map the SIRT1 interaction site in FoxP3. C-terminal deletion at amino acids 278 reduced SIRT1 binding to FoxP3, pointing to amino acids 279 to 337 as a region involved in FoxP3 binding to SIRT1 (FIG. 10B, lanes 2-4). In addition, the N-terminal deletion at amino aids 187 abrogated binding to SIRT1 identifying a second interaction site with SIRT1 at amino acids 133 and 187 of FoxP3 (FIG. 2B, lane 9).
  • To further explore the possibility that SIRT1 deacetylates FoxP3, cells expressing FLAG-FoxP3 were subjected to treatment with nicotinamide, a natural byproduct and feedback inhibitor of the sirtuin deacetylation reaction. Nicotinamide treatment increased acetylation levels of FoxP3 in synergy with the histone acetyltransferase p300 supporting the model that SIRT1 is involved in FoxP3 deacetylation (FIG. 10C, lane 3). Interestingly, nicotinamide-induced hyperacetylation of FoxP3 was more pronounced than hyperacetylation induced by trichostatin A (TSA), a class I and II HDAC inhibitor previously shown to increase FoxP3 acetylation (Li et al., 2007; Tao et al., 2007) (FIG. 10C, lane 2). No synergy was observed when cells were treated with nicotinamide and TSA together (FIG. 10C, lane 4). In contrast, acetylation of another transcription factor, AML1/Runx1 was increased to the same extent by TSA and nicotinamide treatment and both agents synergized to further increase acetylation (FIG. 10D).
  • SIRT1 Destabilizes the FoxP3 Protein Via K31, K262, and K267
  • During these studies of FoxP3 acetylation, noticed an upregulation of protein expression in response to nicotinamide as well as TSA was noticed. To further study the effect of SIRT1 on FoxP3 protein expression, cells were treated with Ex-527, a specific inhibitor of SIRT1. Ex-527 treatment increased FoxP3 protein expression to a similar extent as nicotinamide, supporting the model that SIRT1 deacetylates and destabilizes FoxP3 (FIG. 11A). No effect of Ex-527 or nicotinamide treatment was observed on FoxP3 mRNA levels. In addition, expression of FLAG-AML1/Runx1, which was driven by the same promoter as FoxP3, was unchanged in response to Ex-527 treatment further excluding transcriptional effects of Ex-527 on FoxP3 expression (FIG. 11B). In contrast, treatment with TSA increased both FoxP3 and AML1/Runx1 expression confirming previously described transcriptional effects of TSA on the CMV promoter. This result indicates that inhibition of SIRT1 deacetylase activity by Ex-527 stabilizes FoxP3 expression at the post-transcriptional level.
  • To map the acetylation sites in FoxP3, mass spectrometry was performed on FLAG-FoxP3 isolated from 293 T cells. Flag-FoxP3 was expressed in 293 T cells, immunoprecipitated using anti-Flag agarose, eluted with FLAG peptides and analyzed by mass spectrometry. Peptides recovered in the analysis covered 78-82% of the FoxP3 protein. Three acetylated sites were identified: K31, K262, K267 in mouse FLAG-FoxP3 and the corresponding sites: K31, K263 and K268 in human FLAG-FoxP3 (FIG. 11B). Interestingly, peptides containing acetylated K31 and K262 residues were recovered from samples expressing FLAG-Foxp3 alone, while acetylation of K267 was only detected in samples in which p300 was coexpressed and which were also treated with a combination of TSA and nicotinamide (FIG. 11B). This suggests that K267 is a reversible target of p300 and SIRT1/HDACs while K31 and K262 are more stably acetylated in cells. Interestingly, both K262 and K267 are located close to the C-terminal SIRT1 binding region in FoxP3 (aa 278-337).
  • To determine whether the three newly identified acetylated lysine residues regulate FoxP3 stability, mutants were generated in which each of three lysine residues was substituted to arginines (KR), a conservative mutation that preserves the positive charge of the residue. Expression of FLAG-FoxP3 increased in K262R and K267R mutants suggesting that these residues are involved in regulating FoxP3 protein stability (FIG. 11C, lanes 5 and 7). No such effect was observed when K31 was mutated. In addition, treatment with Ex-527 increased expression of the K31R mutant to comparable levels as the wild type FoxP3 protein (FIG. 11C, lanes 3 and 4). This increase was less visible in K262R and K267 R mutants indicating that these residues are targeted by SIRT1 (FIG. 11C, lanes 6 and 8). Expression of the K262/267R double mutant further increased above levels of the single mutants, and Ex-527 treatment could not further increase the expression of the K262/267R double mutant (FIG. 11C, lanes 9 and 10). Mutation of the triple K31/262/267R mutant (3KR) showed a similar effect as the double mutant (FIG. 11C, lanes 13 and 14).
  • To investigate the effect of SIRT1 inhibition and the three newly identified acetylation sites on FoxP3 protein stability, wild type and 3KR mutant FoxP3 protein expression was measured after treatment with cycloheximide (CHX), a general inhibitor of protein translation. Expression of wild type FoxP3 expression was markedly decreased after 4 h of CHX treatment while treatment with Ex-527 stabilized FoxP3 protein expression at this time point (FIG. 11D, lanes 2 and 5). The 3KR mutant FoxP3 protein mimicked the effect of Ex-527 at the 4 hour-time point, and no further stabilization was observed after Ex-527 treatment (FIG. 11D, lanes 8 and 11). Collectively, these data show that the SIRT1-mediated destabilization of FoxP3 is mediated by the newly identified acetylation sites in FoxP3, mainly K262 and K267.
  • Treatment with Ex-527 Stabilizes FoxP3 Expression in Induced and Natural Tregs
  • To test the effect of Ex-527 on FoxP3 expression in natural FoxP3-expressing cells, Ex-527 was added to naïve CD4+ T cells undergoing differentiation into iTreg cells. The number of FoxP3-expressing cells increased up to 3-fold in the presence of Ex-527 as compared to cells treated with the drug vehicle alone (FIG. 12A). In addition, the mean fluorescent intensity (MFI) of FoxP3 expression was increased up to 2-fold (FIG. 12B). As expected, no effect of Ex-527 on FoxP3 mRNA levels was observed, confirming that Ex-527 promotes FoxP3 expression in iTreg cells at the post-transcriptional level.
  • To test the effect of Ex-527 in natural thymus-derived Treg cells, GFP+ cells were isolated from GFP-Foxp3 knock-in mice, in which green fluorescent protein (GFP) is expressed as a fusion protein with FoxP3. Total CD4+ T cells were isolated from spleen and lymph nodes of these mice, and GFP+ T cells were sorted with >95% purity. GFP+ T cells were expanded by anti-CD3/CD28 beads and IL-2 for 5 days when Ex-527 or dimethylsulfoxide (DMSO) was added. Two days later, 75% of cells were GFP-positive regardless whether Ex-527 was added or not; however, the MFI of GFP was increased in Ex-527-treated cells (FIG. 12B, day 7). 7 days later, only 5% of cells remained GFP-positive in control-treated cells versus 45% in Ex-527-treated cultures (FIG. 12B, day 12). In addition, the MFI of FoxP3 remained higher in Ex-527-treated cells versus control-treated cells (387 versus 263). At day 12, FoxP3 expression in Ex-527-treated cells remained visible by western blot analysis (FIG. 12B). No FoxP3 expression was detected from either GFP-negative cells or cells expanded in the absence of Ex-527 (FIG. 12B). These data underline the negative role of SIRT1 deactylase activity on FoxP3 expression in the natural context of induced or natural Treg cells. Thus, SIRT1 inhibitors, e.g., Ex-527, can be used to maximize ex vivo or in vivo expansion of Tregs for therapeutic purposes.
  • FIGS. 9A and 9B. SIRT1 Plays a Negative Role in iTreg Differentiation
  • (A) Western blot analysis of SIRT1 in mouse CD4+ T cells. Naïve CD4+ T cells isolated from mouse spleen and lymph node were activated with CD3 and CD28 antibodies and differentiated into Th1, Th2 and iTreg cells as described in material and methods. (B) Flow cytometry of iTregs infected with a bicistronic retroviral vector expressing SIRT1 (MSCV-SIRT1-Thy1.1) or control vector (MSCV-Thy1.1). Thy1.1 is a surface marker that is coexpressed to gate on infected cells. Infection efficiencies were similar in SIRT1-expressing or control-infected cells (˜20% Thy1.1+ cells). Shown are the percentage of FoxP3+ cells in the Thy1.1+ population and the mean fluorescence intensity (MFI) of FoxP3 per cell. A and B show one representative experiment of two with the same results.
  • FIGS. 10A-D. SIRT1 Interacts with FoxP3 and Inhibition of SIRT1 Deacetylase Activity Increases Acetylation of FoxP3.
  • (A) Communoprecipitation/western blot analysis of FLAG-tagged FoxP3 and HA-tagged SIRT1 after transfection of corresponding expression vectors or empty vector controls into 293T cells. (B) Communoprecipitation assay of SIRT1 and FoxP3 deletion mutants. A schematic summary of the binding of FoxP3 mutants to SIRT1 is shown. The forkhead box (FH) indicates the DNA binding domain ZnF, zinc finger domain; LZ, leucine zipper. (C) Immunoprecipitation/Western blot analysis of acetylated FLAG-FoxP3 in response to trichostatin A (TSA) or nicotinamide in 293 T cells. An expression vector for the p300 acetyltransferase was cotransfected with FoxP3. (D) The same experiment as in (C) using HA-AML1/Runx1 Immunoprecipitations were performed with either anti-FLAG antibody coupled to agarose (α-FLAG agarose) (C) or anti-hemagglutinin antibody coupled to agarose (α-HA agarose) (D) and western blotting with rabbit polyclonal antibody to acetyl-lysine (α-acetyl lysine) (α-AcK), α-FLAG or α-HA antibodies.
  • FIGS. 11A-D. SIRT1 destabilizes FoxP3 protein through deacetylation of three newly identified acetylation sites
  • (A) Western blot analysis of FLAG-FoxP3 or FLAG-AML1/Runx1in 293 T cells treated with nicotinamide (5 mM), Ex-527 (50 μM) or trichostatin A TSA (400 nM) for 16 hours. (B) Scheme of acetylated lysine residues identified by mass spectrometry using immunoprecipitated FLAG-FoxP3 protein. The murine protein is shown. (C) Western blot analysis of wild type or mutant FLAG-FoxP3 proteins in 293 T cells treated with Ex-527 (50 μM) or DMSO as a control. (D) Western blot analysis of wild type or mutant K31/K262/K267R (3KR) FLAG-FoxP3 in 293 cells treated with cycloheximide (CHX) and/or Ex-527 for indicated times.
  • FIGS. 12A and 12B. Inhibition of SIRT1 activity by Ex-527 promotes differentiation of iTreg cells and FoxP3 expression in nTreg cells.
  • (A) Intracellular flow cytometry (% FoxP3+ cells and MFI) of FoxP3 in induced Tregs treated during differentiation with Ex-527 (50 μM) or DMSO as control. Shown are averages of three independent experiments (mean±SD). (B) Flow cytometry of GFP in natural Treg populations isolated from GFP-FoxP3 knock-in mice, in which GFP is expressed as a fusion protein with FoxP3. GFP+nTregs were expanded by α-CD3/CD28 beads and hIL-2 (2000 U/ml) for 5 days. Cells were further cultured with either DMSO or Ex-527 for additional 2 and 7 days and FoxP3 expression was analyzed as in (A). Western blot analysis of FoxP3 in cell lysates from GFP−, GFP+, and GFP+ cultures expanded as described in the presence of DMSO or EX-527.
  • REFERENCES
    • Chen, L. F., Williams, S. A., Mu, Y., Nakano, H., Duerr, J. M., Buckbinder, L., and Greene, W. C. (2005). NF-kappaB RelA phosphorylation regulates RelA acetylation. Mol Cell Biol 25, 7966-7975.
    • Fu, S., Zhang, N., Yopp, A. C., Chen, D., Mao, M., Chen, D., Zhang, H., Ding, Y., and Bromberg, J. S. (2004). TGF-beta induces Foxp3+ T-regulatory cells from CD4+CD25-precursors. Am J Transplant 4, 1614-1627.
    • Jordan, A., Defechereux, P., and Verdin, E. (2001). The site of HIV-1 integration in the human genome determines basal transcriptional activity and response to Tat transactivation. EMBO J. 20, 1726-1738.
    • Kwon, H. S., Brent, M. M., Getachew, R., Jayakumar, P., Chen, L. F., Schnolzer, M., McBurney, M. W., Marmorstein, R., Greene, W. C., and Ott, M. (2008). Human immunodeficiency virus type 1 Tat protein inhibits the SIRT1 deacetylase and induces T cell hyperactivation. Cell Host Microbe 3, 158-167.
    • Li, B., and Greene, M. I. (2007). FOXP3 actively represses transcription by recruiting the HAT/HDAC complex. Cell Cycle, 6, 1432-1436.
    • Li, B., Samanta, A., Song, X., Iacono, K. T., Bembas, K., Tao, R., Basu, S., Riley, J. L., Hancock, W. W., Shen, Y., et al. (2007). FOXP3 interactions with histone acetyltransferase and class II histone deacetylases are required for repression. Proc Natl Acad Sci USA 104, 4571-4576.
    • Ono, M., Yaguchi, H., Ohkura, N., Kitabayashi, I., Nagamura, Y., Nomura, T., Miyachi, Y., Tsukada, T., and Sakaguchi, S. (2007). Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature 446, 685-689.
    • Pagans, S., Pedal, A., North, B. J., Kaehlcke, K., Marshall, B. L., Don, A., Hetzer-Egger, C., Henklein, P., Frye, R., McBurney, M. W., et al. (2005). SIRT1 Regulates HIV Transcription via Tat Deacetylation. PLoS Biol 3, e41.
    • Tao, R., de Zoeten, E. F., Ozkaynak, E., Chen, C., Wang, L., Porrett, P. M., Li, B., Turka, L. A., Olson, E. N., Greene, M. I., et al. (2007). Deacetylase inhibition promotes the generation and function of regulatory T cells. Nature Med 13, 1299-1307.
    • Zhang, J., Lee, S. M., Shannon, S., Gao, B., Chen, W., Chen, A., Divekar, R., McBurney, M. W., Braley-Mullen, H., Zaghouani, H., and Fang, D. (2009). The type III histone deacetylase Sirtl is essential for maintenance of T cell tolerance in mice. J Clin Invest 119, 3048-3058.
  • While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.

Claims (27)

1.-9. (canceled)
10. A method of modulating activation and differentiation of a CD4+ T cell, the method comprising contacting the CD4+ T cell with a SIRT1 inhibitor.
11. The method of claim 10, wherein the CD4+ T cell is a helper T cell, and wherein contacting results in reduced deacetylation of NF-κB in the helper T cell.
12. The method of claim 11, wherein said contacting takes place in an individual in vivo and wherein said contacting results in increasing in an immune response to an antigen in the individual.
13. The method of claim 12, wherein the individual is a human.
14. The method of claim 12, further comprising administering the antigen to the individual.
15. The method of claim 12, further comprising administering to the individual at least one additional agent that increases an immune response.
16. The method of claim 15, wherein the at least one additional agent is a Toll-like receptor agonist, an immunostimulatory polynucleotide, an immunostimulatory polynucleotide-antigen conjugate, or an immunostimulatory cytokine.
17. The method of claim 10, wherein the CD4+ T cell is a CD4+/CD25+/FoxP3+ T cell, and wherein said contacting results in reduced deacetylation of FoxP3 in the CD4+/CD25+/FoxP3+ T cell.
18. The method of claim 17, wherein said contacting takes place in an individual in vivo and wherein said contacting results in an increase in activity and/or levels of the CD4+/CD25+/FoxP3+ T cell in the individual.
19. The method of claim 18, wherein said increase in activity and/or levels of the CD4+/CD25+/FoxP3+ T cell in the individual is effective to treat an autoimmune disorder or to reduce graft rejection.
20. The method of claim 18, wherein the individual is a human.
21. The method of claim 10, wherein the SIRT1 inhibitor is 7-chloro-1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; 6-bromo-2,3,4,9-tetrahydro-1H-carbazole-2-carboxylic acid amide; 1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; or 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid (5-chloro-pyridin-2-yl)-amide.
22. The method of claim 12, wherein the SIRT1 inhibitor is administered orally.
23. The method of claim 18, wherein the SIRT1 inhibitor is administered orally.
24. A method of treating an autoimmune disorder in an individual, the method comprising administering to the individual an effective amount of a SIRT1 inhibitor.
25. The method of claim 24, wherein the individual is a human.
26. The method of claim 24, wherein the SIRT1 inhibitor is 7-chloro-1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; 6-bromo-2,3,4,9-tetrahydro-1H-carbazole-2-carboxylic acid amide; 1,2,3,4-tetrahydro-cyclopenta[b]indole-3-carboxylic acid amide; or 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid (5-chloro-pyridin-2-yl)-amide.
27. The method of claim 24, wherein the SIRT1 inhibitor is administered orally.
28. The method of claim 24, wherein the autoimmune disorder is multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Type 1 diabetes, or autoimmune hemolytic anemia.
29. The method of claim 24, wherein the SIRT1 inhibitor is selected from:
Figure US20140030295A1-20140130-C00665
Figure US20140030295A1-20140130-C00666
Figure US20140030295A1-20140130-C00667
30. The method of claim 24, wherein the SIRT1 inhibitor is selected from:
Figure US20140030295A1-20140130-C00668
31. The method of claim 24, further comprising administering to the individual at least one additional therapeutic agent.
32. The method of claim 31, wherein the at least one additional therapeutic agent is selected from a corticosteroid drug, a non-steroidal anti-inflammatory drug, and an immunosuppressant drug.
33. The method of claim 31, wherein the autoimmune disorder is Type 1 diabetes, and the at least one additional therapeutic agent is insulin or an insulin analog.
34. The method of claim 31, wherein the autoimmune disorder is multiple sclerosis, and the at least one additional therapeutic agent is interferon-alpha.
35. The method of claim 24, wherein the SIRT1 inhibitor is formulated with an enteric-soluble coating material.
US13/717,041 2008-02-07 2012-12-17 Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response Abandoned US20140030295A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/717,041 US20140030295A1 (en) 2008-02-07 2012-12-17 Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US2699708P 2008-02-07 2008-02-07
PCT/US2009/000761 WO2009099643A1 (en) 2008-02-07 2009-02-05 Use of sirt1 activators or inhibitors to modulate an immune response
US12/838,247 US8748464B2 (en) 2008-02-07 2010-07-16 Use of SIRT1 activators or inhibitors to modulate an immune response
US13/717,041 US20140030295A1 (en) 2008-02-07 2012-12-17 Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/838,247 Division US8748464B2 (en) 2008-02-07 2010-07-16 Use of SIRT1 activators or inhibitors to modulate an immune response

Publications (1)

Publication Number Publication Date
US20140030295A1 true US20140030295A1 (en) 2014-01-30

Family

ID=40952400

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/838,247 Active 2030-07-09 US8748464B2 (en) 2008-02-07 2010-07-16 Use of SIRT1 activators or inhibitors to modulate an immune response
US13/717,041 Abandoned US20140030295A1 (en) 2008-02-07 2012-12-17 Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/838,247 Active 2030-07-09 US8748464B2 (en) 2008-02-07 2010-07-16 Use of SIRT1 activators or inhibitors to modulate an immune response

Country Status (2)

Country Link
US (2) US8748464B2 (en)
WO (1) WO2009099643A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017123161A1 (en) * 2016-01-15 2017-07-20 Agency For Science, Technology And Research Inhibition of intracellular growth of mycobacterium species and its applications

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012012737A2 (en) * 2010-07-23 2012-01-26 The University Of Toledo Stable tregs and related materials and methods
WO2019099560A1 (en) 2017-11-14 2019-05-23 The Schepens Eye Research Institute, Inc. Runx1 inhibition for treatment of proliferative vitreoretinopathy and conditions associated with epithelial to mesenchymal transition
CN115916218A (en) * 2020-04-23 2023-04-04 斯格本斯眼科研究所有限公司 Methods and materials for treating fibrosis
CN116426483B (en) * 2021-12-30 2024-02-23 南京紫珑生物科技有限公司 Application of CD258 protein in immunotherapy

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040091936A1 (en) * 2002-05-24 2004-05-13 Michael West Bank of stem cells for producing cells for transplantation having HLA antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
WO2007047604A2 (en) * 2005-10-18 2007-04-26 Elixir Pharmaceuticals, Inc. Sirt1 inhibition
US7387271B2 (en) * 2002-12-30 2008-06-17 3M Innovative Properties Company Immunostimulatory combinations

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7223398B1 (en) * 1999-11-15 2007-05-29 Dynavax Technologies Corporation Immunomodulatory compositions containing an immunostimulatory sequence linked to antigen and methods of use thereof
US20070105109A1 (en) * 2003-07-02 2007-05-10 Geesaman Bard J Sirt1 and genetic disorders
EP1677794B1 (en) * 2003-09-12 2014-12-03 Elixir Pharmaceuticals, Inc. Methods of treating disorder
AU2005207029B2 (en) 2004-01-20 2011-09-01 Brigham Young University Novel sirtuin activating compounds and methods for making the same
AU2006278504B2 (en) 2005-08-04 2013-01-17 Sirtris Pharmaceuticals, Inc. Imidazopyridine derivatives as sirtuin modulating agents
WO2007084775A2 (en) 2006-01-20 2007-07-26 The Trustees Of The University Of Pennsylvania Compositions and methods for modulation of suppressor t cell activation
TW200918542A (en) 2007-06-20 2009-05-01 Sirtris Pharmaceuticals Inc Sirtuin modulating compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040091936A1 (en) * 2002-05-24 2004-05-13 Michael West Bank of stem cells for producing cells for transplantation having HLA antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
US7387271B2 (en) * 2002-12-30 2008-06-17 3M Innovative Properties Company Immunostimulatory combinations
WO2007047604A2 (en) * 2005-10-18 2007-04-26 Elixir Pharmaceuticals, Inc. Sirt1 inhibition

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Ansel, An epigenetic view of helper T cell differentiation, Nature Immunology, 2003, 4(7), pp. 616-623. *
Cheng, Developmental defects and p53 hyperacetylation in Sir2 homolog (SIRT1)-deficient mice, PNAS, 2003, 100 (19), pp. 10794-10799. *
Danziger et al., Automated Site-directed Drug Design: A General Algorithm for Knowledge Acquisition about Hydrogen-Bonding Regions at Protein Surfaces, Mar.22, 1989, The Royal Society, Proceedings of the Royal Society of London.Series B, Biological Sciences, Vol.236, No.1283, p.101-113. *
Jindal, Formulation and evaluation of insulin enteric microspheres for oral drug delivery, Acta Pharmaceutica Sciencia, 2009, 51, pp. 121-127. *
Rabinovitch Roles of cytokines in the pathogenesis and therapy of type 1 diabetes, Cell. Biochem. Biophys., 2007, 48, pp. 159-163 *
Tao, Deacetylase inhibition promotes the generation and function of regulatory T cells, Nature Medicine, 2007, 13(11), pp. 1299-1307. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017123161A1 (en) * 2016-01-15 2017-07-20 Agency For Science, Technology And Research Inhibition of intracellular growth of mycobacterium species and its applications

Also Published As

Publication number Publication date
US20100330114A1 (en) 2010-12-30
WO2009099643A1 (en) 2009-08-13
US8748464B2 (en) 2014-06-10

Similar Documents

Publication Publication Date Title
US11298328B2 (en) Sensorineural hair cell differentiation
McGaha et al. Amino acid catabolism: a pivotal regulator of innate and adaptive immunity
US20140030295A1 (en) Use of Sirt1 Activators or Inhibitors to Modulate an Immune Response
WO2007084775A9 (en) Compositions and methods for modulation of suppressor t cell activation
US10653679B2 (en) Compositions and methods for inhibiting bacterial growth
WO2014189648A1 (en) Methods and compositions for treatment of hiv infection
AU2012275190B2 (en) Compositions, methods and kits for treating leukemia
AU2020236937B2 (en) CD40L compositions and methods for tunable regulation
HUE031652T2 (en) Novel composition for the treatment of cystic fibrosis
Casalino-Matsuda et al. Hypercapnia suppresses macrophage antiviral activity and increases mortality of influenza A infection via Akt1
US20150099012A1 (en) Methods for activating retrovirus in latent infected cells, and compounds for use therein
JP2014528959A (en) Methods and pharmaceutical compositions for the treatment of Th2-mediated diseases
US20190247367A1 (en) Treatment of infectious diseases
US11191767B2 (en) Pharmaceutical composition for stimulating stem cell division and suppressing bacterial virulence
JP2006117536A (en) Medicine for inducing hair cell of internal ear
CN111629716A (en) Methods and compositions for treating inflammation
Occhigrossi et al. The STING/TBK1/IRF3/IFN type I pathway is defective in cystic fibrosis
US20230226095A1 (en) Methods and compositions for treating coronavirus infectious disease
US20240173276A1 (en) Methods and compositions for inhibiting formation of the hiv latent reservoir
RU2819782C2 (en) Methods of treating virus-associated forms of cancer with histone deacetylase inhibitors
EP3954378A1 (en) Compositions comprising t-cells for topical administration to the lung
WO2023150507A2 (en) 7dw8-5 treatment for covid-19 and other virus-induced respiratory infections
Hsieh et al. Equilibrative Nucleoside Transporter 3 is an IFN-stimulated Gene that Facilitates Viral Genome Release
WO2022047182A1 (en) High dose nicotinamide adenine dinucleotide (nad) precursor regimens for reduction of inflammation in human patients with preexisting inflammation
Yerinde Characterization of the Role of Histone Deacetylase 7 in CD8+ T Cell Dependent Anti-tumor Immune Responses and T Cell Homeostasis

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE J. DAVID GLADSTONE INSTITUTES, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIM, HYUNGWOOK;OTT, MELANIE;VERDIN, ERIC M.;AND OTHERS;SIGNING DATES FROM 20130107 TO 20130124;REEL/FRAME:029730/0804

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:J. DAVID GLADSTONE INSTITUTES;REEL/FRAME:029872/0029

Effective date: 20130107

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: THE J. DAVID GLADSTONE INSTITUTES, A TESTAMENTARY TRUST ESTABLISHED UNDER THE WILL OF J. DAVID GLADSTONE, CALIFORNIA

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE NAME TO THE J. DAVID GLADSTONE INSTITUTES, A TESTAMENTARY TRUST ESTABLISHED UNDER THE WILL OF J. DAVID GLADSTONE PREVIOUSLY RECORDED ON REEL 029730 FRAME 0804. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:LIM, HYUNGWOOK;OTT, MELANIE;VERDIN, ERIC M.;AND OTHERS;SIGNING DATES FROM 20130107 TO 20130124;REEL/FRAME:052425/0866

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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