WO2019106427A2 - Procédé de criblage d'agents d'inversion de latence du vih-1 - Google Patents

Procédé de criblage d'agents d'inversion de latence du vih-1 Download PDF

Info

Publication number
WO2019106427A2
WO2019106427A2 PCT/IB2018/001456 IB2018001456W WO2019106427A2 WO 2019106427 A2 WO2019106427 A2 WO 2019106427A2 IB 2018001456 W IB2018001456 W IB 2018001456W WO 2019106427 A2 WO2019106427 A2 WO 2019106427A2
Authority
WO
WIPO (PCT)
Prior art keywords
hiv
cells
inhibitor
lra
fraction
Prior art date
Application number
PCT/IB2018/001456
Other languages
English (en)
Other versions
WO2019106427A3 (fr
Inventor
Privada Institut De Recerca De La Sida-Caixa Fundacio
Julia GARCIA PRADO
Alba RUIZ DE ANDRES
Original Assignee
Fundacio Privada Inst De Recerca De La Sida Caixa
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 Fundacio Privada Inst De Recerca De La Sida Caixa filed Critical Fundacio Privada Inst De Recerca De La Sida Caixa
Publication of WO2019106427A2 publication Critical patent/WO2019106427A2/fr
Publication of WO2019106427A3 publication Critical patent/WO2019106427A3/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70514CD4

Definitions

  • the present invention relates to a method for screening HIV-l latency reversing agents (LRA).
  • LRA HIV-l latency reversing agents
  • HIV infection is one of the major threats to global human health. It is estimated that more than 78 million people worldwide have been infected by the human immunodeficiency virus since 1981. Nearly half of these infected individuals have died of the resultant Acquired Immunodeficiency Syndrome (AIDS) during the same time frame. See UNAIDS, htp://www.unaids.ofg/, November 2017.
  • AIDS Acquired Immunodeficiency Syndrome
  • ART antiretroviral therapy
  • HDACi histone deacetylases inhibitors
  • PLCm protein kinase C
  • cytotoxic T-lymphocytes have been put forward as candidates to elicit this function due to their in vitro activity in eliminating reactivated cells and their key role in the control of the reservoir in natural HIV-l elite controllers. See Shan L, et al, Immunity 2012; 36(3):49l-50l, Descours B, et al, Clin Infect Dis 20l2;54(l0): 1495-503 and Noel N, et al, J Virol 2016; 90(13):6148-6158. Nonetheless, the direct effect of HDACi and PKCm in CTL activity should be taken into account to identify the therapeutic window of activity between latency-reversing agents and CTL clearance of reactivated cells.
  • the present invention is directed to a method for screening HIV-l latency reversing agents (LRA).
  • the method comprises the steps of (a) culturing in vitro a fraction of CD4+ T cells obtained from an HIV-infected subject with at least one latency reversing agent (LRA) for at least 48 hours, (b) co-culturing in vitro the fraction of CD4+ T cells with cytotoxic T-lymphocytes (CTL) obtained from the HIV-infected subject and (c) counting the killed CD4+ T cells in the fraction.
  • Said screening could be useful in the treatment and prevention of HIV-l infection and AIDS, such as by assisting in the optimization of ART and HAART protocols for HIV-infected subjects.
  • a method for screening latency reversing agents which comprises:
  • the LRA comprises a protein kinase C (PKC) modulator, a histone deacetylase (HDAC) inhibitor or a combination thereof.
  • PLC protein kinase C
  • HDAC histone deacetylase
  • HDAC inhibitor comprises panobinostat, romidepsin, trichostatin A and vorinostat.
  • the present invention refers to a kit comprising at least one of the LRA of the invention.
  • Fig. 1 Representation of the shock and kill RELI model. Briefly, the HIV-l permissive U937 cell line, transfected with the HLA-B* 27:05 gene, was infected for three days with a NL43 strain carrying a reporter GFP gene. The GFP negative population was then sorted and cultured in the presence of the protease inhibitor RTV to prevent viral spread and favor the establishment of an enriched latently infected state. RELI cells were reactivated with LRA in the presence of the integrase inhibitor RAL before being extensively washed and cultured in the presence or absence of HLA-class I matched HIV-specific CTL and RAL. 20 hours after coculture, cells were stained and analyzed by flow cytometry to evaluate HIV- 1 reactivation (shock) and CTL-mediated elimination of HIV-reactivated RELI cells (kill).
  • Fig. 2 Representative dot plots showing intracellular p24 levels from live RELI cells reactivated with HDACi combined or not with the PKC agonist bryostatin.
  • Fig. 3 Representative dot plots showing intracellular p24 and GFP levels from live RELI cells reactivated with LRA.
  • Fig. 4 Correlation between p24 expression and GFP expression in RELI cells after LRA treatment.
  • the line indicates the fit of the data to a linear regression.
  • the Spearman correlation coefficient (rho) and the two-tailed p-value are shown.
  • the graph represents the mean ⁇ SEM of five independent experiments performed in duplicates.
  • Fig. 5 HIV-l reactivation measured as percentage of intracellular p24 levels and GFP expression.
  • the graph represents the mean ⁇ SD of two replicates from one representative experiment.
  • Fig. 6 HIV-l fold induction measured as the relative p24 expression level compared to untreated condition.
  • the graph represents the mean ⁇ SEM of five independent experiments performed in duplicates. The p-values were calculated using the one-sample t test. Only significant values are shown in the figure (p ⁇ 0.05 *, p ⁇ 0.005 **).
  • administering includes any method of delivering to a subject a cellular composition according to the present invention.
  • AIDS refers to the symptomatic phase of HIV infection, and includes both Acquired Immune Deficiency Syndrome (commonly known as AIDS) and “ARC,” or AIDS-Related Complex. See Adler M, et al., Brit. Med. J. 1987; 294: 1145-1147.
  • the immunological and clinical manifestations of AIDS are well known in the art and include, for example, opportunistic infections and cancers resulting from immune deficiency.
  • antiretroviral therapy refers to the administration of one or more antiretroviral drugs (i.e. HIV antiretrovirals) to inhibit the replication of HIV.
  • ART involves the administration of at least one antiretroviral agent (or, commonly, a cocktail of antiretrovirals) such as nucleoside reverse transcriptase inhibitor (e.g. zidovudine (AZT, lamivudine (3TC) and abacavir), non-nucleoside reverse transcriptase inhibitor (e.g. nevirapine and efavirenz) and protease inhibitor (e.g. indinavir, ritonavir and lopinavir).
  • nucleoside reverse transcriptase inhibitor e.g. zidovudine (AZT, lamivudine (3TC) and abacavir
  • non-nucleoside reverse transcriptase inhibitor e.g. nevirapine and efavirenz
  • protease inhibitor
  • HAART Highly Active Antiretroviral Therapy
  • HAART refers to treatment regimens designed to suppress aggressively HIV replication and disease progression.
  • HAART usually consists of three or more different drugs, such as, for example, two nucleoside reverse transcriptase inhibitors and a protease inhibitor.
  • histone deacetylase inhibitor refers to a compound that inhibits histone deacetylase enzymes.
  • Histone deacetylases remove acetyl groups from lysine residues on histones during this process, thus allowing histones to wrap DNA more tightly.
  • HDAC inhibitors prevent deacetylation and, therefore, affect gene expression. Examples of HDAC inhibitors include, but are not limited to, apicidin, abexinostat (i.e. PCI-24781), belinostat (i.e. Beleodaq®), BRD 4354, dacinostat (i.e.
  • LAQ- 824 depudecin, droxinostat, entinostat (i.e. SNDX-275, MS-275), givinostat (i.e. gavinostat, ITF 2357), KD 5170, LMK 235, M 344, MC 1568, MC 1742, MI 192, mocetinostat (i.e. MGCD-0103), NCH 51 (i.e. PTACH), NSC 3852 (i.e. 5-nitroso-8-quinolinol), niltubacin (i.e. MAZ-1391), oxamflatin (i.e. metacept 3), panobinostat (i.e.
  • Farydak® PCI 34051, pracinostat (i.e. SB 939), pyroxamide, quisinostat (i.e. JNJ-26481585), resminostat (i.e. 4SC- 201, RAS2410), RG2833 (i.e. RGFP109), RGFP966, Ricinus communis toxin (i.e. RC- Toxin), rocilinostat (i.e. ACY-1215), romidepsin (i.e. Istodax®, FK 228), scriptaid, sodium butyrate, sodium 4-phenylbutyrate, suberohydroxamic acid (i.e.
  • SBHA N,N'- dihydroxyoctanediamide
  • C-H 106 tacedinaline (i.e. N-acetyldinaline, Cl 994), TCS HDAC6 20b, TMP269, trichostatin A, tubacin, UF 010 (i.e. 4-bromo-N'- butylbenzohydrazide), valproic acid, and vorinostat (i.e. Zolinza®, SAHA).
  • HIV include HIV-l and HIV-2, SHIV and SIV.“HIV- 1” means the human immunodeficiency virus type-l. HIV-l includes, but is not limited to, extracellular virus particles and the forms of HIV-l associated with HIV-l infected cells. The HIV-l virus may represent any of the known major subtypes (Classes A, B, C, D E, F, G and H) or outlying subtype (Group O) including laboratory strains and primary isolates.“HIV-2” means the human immunodeficiency virus type-2. HIV-2 includes, but is not limited to, extracellular virus particles and the forms of HIV-2 associated with HIV-2 infected cells.
  • SIV refers to simian immunodeficiency virus which is an HIV-like virus that infects monkeys, chimpanzees, and other nonhuman primates. SIV includes, but is not limited to, extracellular virus particles and the forms of SIV associated with SIV infected cells.
  • HIV infection refers to indications of the presence of the HIV virus in an individual including asymptomatic seropositivity, AIDS-related complex (ARC), and acquired immunodeficiency syndrome (AIDS).
  • ARC AIDS-related complex
  • AIDS acquired immunodeficiency syndrome
  • Integrase inhibitor refers to a compound that can block the action of integrase, a viral enzyme that inserts the viral genome into the DNA of the host cell. Integrase inhibitors include integrase strand transfer inhibitors (INSTIs) and integrase binding inhibitors (INBIs).
  • An“HIV integrase inhibitor” is an integrase inhibitor used in the treatment and prevention of HIV infection and AIDS. Examples of HIV integrase inhibitors include, but are not limited to, aurintricarboxylic acid, bictegravir (i.e. GS-9883), cabotegravir (i.e.
  • GSK1265744, GSK744) caffeic acid phenethyl ester, curcumin, chicoric acid, 3,5- dicaffeoylquinic acid, 4-(2,3-dihydropyrano[4,3,2-de]quinolin-7-yl)-a-(l, 1 -dimethyl ethoxy)- 2-methyl-3-quinolineacetic acid (i.e. BI 224436), dolutegravir (i.e. Tivicay®), elvitegravir (i.e. Vitekta®), globoidnan A, MK-2048, quercetin, raltegravir (i.e. Isentress®), tyrphostin, and their derivatives.
  • dolutegravir i.e. Tivicay®
  • elvitegravir i.e. Vitekta®
  • globoidnan A MK-2048
  • quercetin quercetin
  • latency reversing agents include, but not are not limited to, protein kinase C modulators (PKCm), bromo and external (BET) bromodomain inhibitors, histone deacetylase inhibitors (HDACi), acetaldehyde dehydrogenase inhibitors (e.g. disulfiram), activators of nuclear factor kappa-light chain-enhancer of activated B cells (NF- KB), and activators of the AKT pathway.
  • PICm protein kinase C modulators
  • BET bromo and external bromodomain inhibitors
  • HDACi histone deacetylase inhibitors
  • acetaldehyde dehydrogenase inhibitors e.g. disulfiram
  • activators of nuclear factor kappa-light chain-enhancer of activated B cells NF- KB
  • activators of the AKT pathway activators of the AKT pathway.
  • BET inhibitors include, but are not limited to, CPI 203,
  • HIV protease inhibitor refers to a compound that can prevent viral replication by binding selectively to viral proteases and, therefore, blocks the cleavage of protein precursors that are necessary for making infectious viral particles.
  • An“HIV protease inhibitor” is a protease inhibitor used in the treatment and prevention of HIV infection and AIDS.
  • HIV protease inhibitors include, but are not limited to, amprenavir (i.e. Agenerase®), atazanavir (i.e. Reyataz®), darunavir (i.e. Prezista®), fosamprenavir (i.e. Lexiva®, Telzir®), indinavir (i.e.
  • Crixivan® lopinavir (i.e. Kaletra®), nelfmavir (i.e. Viracept®), ritonavir (i.e. Norvir®), saquinavir (i.e. Invirase®, Fortovase®), and tipranavir (Aptivus®).
  • PKC modulator refers to a compound that affects the activity of a protein kinase C (“PKC”) enzyme.
  • PKCs are enzymes that regulate the function of other proteins through the phosphorylation of hydroxyl groups located in the serine and threonine residues of said proteins.
  • PKC modulators can be antagonists (i.e. inhibitors), agonists (i.e. activators) or both. Examples of PKC antagonists include, but are not limited to, bisindolylmaleimide 1 (i.e.
  • BIM-l, GF 109203X and its related compounds BIM-2, BIM-3 and BIM-8, [Ala 107 J- MBP (104-118), [Ala 113 ]-MBP (104-118), C-l, calphostin C (i.e. UCN 1028C, PKF 115584), CGP 53353, chelerythrine, D-erythro-sphingosine, dihydrosphingosine (i.e. sphinganine), enzastaurin (i.e. LY317615), [Glu 27 ]-PKC (19-36), Go 6976, Go 6983, gossypol, melittin, myricitrin (i.e.
  • PKC agonists include, but are not limited to, bryostatin A (i.e. bryostatin 1, NSC 339555) and its analogs bryostatin 2 and bryostatin 3, diacyl glycerol (DAG) mimics, FR 236924 (i.e. DCP- LA), ingenol mebutate (i.e. Picato®, ingenol-3-angelate, PEP005) and its analogs, phorbol 12, 13 -dibutyrate (i.e. PDbu), phorbol l2-myristate l3-acetate (i.e. PMA), prostratin, pseudo RACK1, SC-10, and SC-9.
  • bryostatin A i.e. bryostatin 1, NSC 339555
  • DAG diacyl glycerol
  • FR 236924 i.e. DCP- LA
  • ingenol mebutate i.e. Picato®, ingenol-3
  • subject refers to an individual or animal, such as a human, a nonhuman primate (e.g. chimpanzees and other apes and monkey species); farm animals, such as birds, fish, cattle, sheep, pigs, goats and horses; domestic mammals, such as dogs and cats; laboratory animals including rodents, such as mice, rats and guinea pigs.
  • a nonhuman primate e.g. chimpanzees and other apes and monkey species
  • farm animals such as birds, fish, cattle, sheep, pigs, goats and horses
  • domestic mammals such as dogs and cats
  • laboratory animals including rodents, such as mice, rats and guinea pigs.
  • rodents such as mice, rats and guinea pigs.
  • the term“subject” encompasses an embryo and a fetus. In a preferred embodiment, the subject is a human.
  • control of the disease progression refers to mean the beneficial or desired clinical results that include, but are not limited to, reduction of the symptoms, reduction of the duration of the disease, stabilization of pathological states (specifically to avoid additional deterioration), delaying the progression of the disease, improving the pathological state and remission (both partial and total).
  • the control of progression of the disease also involves an extension of survival compared with the expected survival if treatment was not applied.
  • the terms “treat” and“treatment” refer specifically to stopping or slowing the infection and destruction of healthy CD4+ T cells in an HIV-infected subject. It also refers to the stopping and slowing of the onset of symptoms of the acquired immunodeficiency disease such as extreme low CD4+ T cell count and repeated infections by opportunistic pathogens.
  • Beneficial or desired clinical results include, but are not limited to, an increase in absolute naive CD4+ T cell count (range 10-3520), an increase in the percentage of CD4+ T cell over total circulating immune cells (range 1-50%), or an increase in CD4+ T cell count as a percentage of normal CD4+ T cell count in an uninfected subject (range 1-161%).“Treatment” can also mean prolonging survival of the infected subject as compared to expected survival if the subject does not receive any HIV targeted treatment.
  • the present invention refers to a method for screening latency reversing agents (LRA) which comprises the steps of (a) culturing in vitro a fraction of CD4+ T cells obtained from a HIV-infected subject with at least one latency reversing agent (LRA) for at least 48 hours, (b) co-culturing in vitro the fraction of CD4+ T cells with cytotoxic T- lymphocytes (CTL) obtained from the HIV-infected subject and (c) counting the killed CD4+ T cells in the fraction.
  • LRA latency reversing agents
  • the LRA comprises a protein kinase C (PKC) modulator, a histone deacetylase (HD AC) inhibitor or a combination thereof.
  • PLC protein kinase C
  • HD AC histone deacetylase
  • the PKC modulator is a PKC agonist.
  • the PKC agonist comprises bryostatin A, bryostatin 2, bryostatin 3, DAG mimics, FR 236924, ingenol mebutate, phorbol 12, 13 -dibutyrate, phorbol l2-myristate l3-acetate, prostratin, pseudo RACK1, SC-10, and SC-9 or a combination thereof. More preferably, the PKC agonist comprises bryostatin A.
  • the HDAC inhibitor comprises apicidin, abexinostat, belinostat, BRD 4354, dacinostat, depudecin, droxinostat, entinostat, givinostat, KD 5170, LMK 235, M 344, MC 1568, MC 1742, MI 192, mocetinostat, NCH 51, 5-nitroso-8- quinolinol, niltubacin, oxamflatin, panobinostat, PCI 34051, pracinostat, pyroxamide, quisinostat, resminostat, RG2833, RGFP966, Ricinus communis toxin, rocilinostat, romidepsin, scriptaid, sodium butyrate, sodium 4-phenylbutyrate, suberohydroxamic acid, C- H 106, tacedinaline, TCS HDAC6 20b, TMP269, trichostatin A, tub
  • a combination of a PKC modulator and a HDAC inhibitor is employed as LRA.
  • a combination of a PKC agonist and a HDAC inhibitor is used.
  • a combination of a PKC antagonist and a HDAC inhibitor is employed.
  • a combination of: a PKC agonist and panobinostat, a PKC agonist and romidepsin, a PKC agonist and trichostatin A, or a PKC agonist and vorinostat is employed.
  • a combination of: bryostatin A and panobinostat, bryostatin A and romidepsin, bryostatin A and trichostatin A, or bryostatin A and vorinostat is used.
  • an HIV protease inhibitor is added to the fraction of CD4+ T cells of step (a) before culturing said fraction with the LRA.
  • amprenavir, atazanavir, darunavir, fosamprenavir, indinavir, lopinavir, nelfmavir, ritonavir, saquinavir, and tipranavir or a combination thereof is used as protease inhibitor. More preferably, ritonavir is employed as protease inhibitor.
  • the LRA and HIV protease inhibitor of the method of the invention comprise, respectively, (a) a combination of a PKC agonist and panobinostat and (b) ritonavir, (a) a combination of a PKC agonist and romidepsin and (b) ritonavir, (a) a combination of a PKC agonist and trichostatin A and (b) ritonavir, or (a) a combination of a PKC agonist and vorinostat and (b) ritonavir.
  • the LRA and HIV protease inhibitor of the method comprise, respectively, (a) a combination of bryostatin A and panobinostat and
  • ritonavir (a) a combination of bryostatin A and romidepsin and (b) ritonavir, (a) a combination of bryostatin A and trichostatin A and (b) ritonavir, or (a) a combination of bryostatin A and vorinostat and (b) ritonavir.
  • an HIV integrase inhibitor is added to the fraction of CD4+ T cells of step (a) when culturing said fraction with the LRA.
  • cabotegravir, dolutegravir, elvitegravir, and raltegravir or a combination thereof is used as integrase inhibitor. More preferably, raltegravir is employed as integrase inhibitor.
  • the LRA and HIV integrase inhibitor of the method of the invention comprise, respectively, (a) a combination of a PKC agonist and panobinostat and
  • raltegravir (a) a combination of a PKC agonist and romidepsin and (c) raltegravir, (a) a combination of a PKC agonist and trichostatin A and (c) raltegravir, or (a) a combination of a PKC agonist and vorinostat and (c) raltegravir.
  • the LRA and HIV integrase inhibitor of the method comprise, respectively, (a) a combination of bryostatin A and panobinostat and (c) raltegravir, (a) a combination of bryostatin A and romidepsin and (c) raltegravir, (a) a combination of bryostatin A and trichostatin A and (c) raltegravir, or (a) a combination of bryostatin A and vorinostat and (c) raltegravir.
  • the LRA, HIV protease inhibitor and HIV integrase inhibitor of the method of the invention comprise, respectively, (a) a combination of a PKC agonist and panobinostat, (b) ritonavir, and (c) raltegravir, (a) a combination of a PKC agonist and romidepsin, (b) ritonavir, and (c) raltegravir, (a) a combination of a PKC agonist and trichostatin A, (b) ritonavir, and (c) raltegravir, or (a) a combination of a PKC agonist and vorinostat, (b) ritonavir, and (c) raltegravir.
  • the LRA, HIV protease inhibitor and HIV integrase inhibitor of the method comprise, respectively, (a) a combination of bryostatin A and panobinostat, (b) ritonavir, and (c) raltegravir, (a) a combination of bryostatin A and romidepsin, (b) ritonavir, and (c) raltegravir, (a) a combination of bryostatin A and trichostatin A, (b) ritonavir, and (c) raltegravir, or (a) a combination of bryostatin A and vorinostat, (b) ritonavir, and (c) raltegravir.
  • kits comprising at least one of the LRA, HIV protease inhibitors or HIV integrase inhibitors of the invention.
  • the kit comprises a combination of at least one PKC modulator and at least one HD AC inhibitor according to the invention. More preferably, the kit comprises a combination of at least one PKC agonist and at least one HDAC inhibitor.
  • the kit comprises (a) a combination of at least one PKC agonist and at least one HDAC inhibitor and (b) at least one HIV protease inhibitor.
  • the kit comprises (a) a combination of at least one PKC agonist and at least one HDAC inhibitor and (c) at least one HIV integrase inhibitor.
  • the kit comprises (a) a combination of at least one PKC agonist and at least one HDAC inhibitor, (b) at least one HIV protease inhibitor, and (c) at least one HIV integrase inhibitor.
  • the kit may include one or more PKC agonists, HDAC inhibitors, HIV protease inhibitors, and HIV integrase inhibitors or combinations thereof.
  • kits of the invention may be optionally packed in suitable containers and be labeled for screening LRA.
  • the components of the kits may be stored in unit or multi-dose containers as an aqueous, preferably sterile, solution or as a lyophilized, preferably sterile, formulation for reconstitution.
  • the containers may be formed from a variety of materials such as glass or plastic and may have a sterile access port (e.g. the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the kits may further comprise more containers comprising a pharmaceutically acceptable carrier.
  • kits may further include other materials desirable from a commercial and user standpoint, including, but not limited to, buffers, diluents, filters, needles, syringes, culture medium for one or more of the suitable cells or other active agents used in the method of the invention.
  • the kits can contain instructions customarily included in commercial packages of diagnostic and screening products that contain information, for example, about the usage, dosage, manufacture, administration, contraindications or warnings concerning the use of such diagnostic and screening products.
  • HLA-B*2705 expressing cells were obtained from the University of Oxford (Oxford, GB). HLA-B*2705 restricted cytotoxic T-lymphocytes (CTL) specific for the recognition of HIV-I epitopes KKlO (KRWIILGLNK) and KY9 (KRKGGIGGY) and KF11 (KAFSPEVIPMF) were prepared following protocols known in the art. See Payne R, el al ., J Virol 2010; 84(20): 10543-10557 and Kloverpris H, et al, J Virol 2013; 87(19): 10889- 10894.
  • CTL cytotoxic T-lymphocytes
  • R10 Prior to infection, the U937-HLA-B*B2705 expressing cells were cultured since day - 3 in a R10 + geneticin 0.5 mg/mL (G418, Cat. No. 10131027, Thermo Fisher Scientific Inc., Waltham, MA, USA) culture medium. A final culture concentration of 0.8-1 M/mL was reached.
  • R10 is composed of RPMI 1640 medium (Cat. No. 21875, Thermo Fisher Scientific Inc., Waltham, MA, USA), penicillin-streptomycin (5,000 UI/mL, Cat. No. 15070063, Thermo Fisher Scientific Inc., Waltham, MA, USA) and 10% fetal bovine serum (FBS, Gibco, Thermo Fisher Scientific Inc., Waltham, MA, USA).
  • the magnetofection plate was then incubated. 0.8 M cells were added to 24-well plates in a volume of 300 pL R10. The virus was mixed with the VM (100 pL NL43-GFP: 4 pL VM). The mixture was incubated for 15 minutes at room temperature. Viral stocks from 1M TCID50/mL were preferably used. The magnetized virus was pipetted carefully into the target wells, making sure of not disturbing the cell layer at the bottom of the wells.
  • the 24-well plates were placed on the magnetofection plates making sure that the magnets were properly lined up. The plates were then incubated at 37°C for 40 minutes. Afterwards, the infected cells were harvested, re-suspended in PBS IX (Cat. No. 10010023, Gibco, Thermo Fisher Scientific Inc., Waltham, MA, USA) and transferred to a microcentrifuge tube. The plates were washed with 1 mL of PBS IX to make sure that all cells were harvested. The cells were subsequently washed twice with PBS IX and were centrifuged at 400 g for 5 minutes.
  • PBS IX Cat. No. 10010023, Gibco, Thermo Fisher Scientific Inc., Waltham, MA, USA
  • the infected cells were placed in 24-well plates at a final concentration of 0.8 M/mL in 1 mL of R10 + geneticin 0.5 mg/mL for 72 hours.
  • the culture was checked at 24 and 48 hours after infection to monitor cellular growth.
  • Fresh new media i.e. around 1 mL was added to the wells if necessary.
  • the frequency of GFP positive cells was measured by flow cytometry after 72 hours of infection.
  • the cells were counted using microbeads.
  • a 10-20% range of GFP-expressing cells was employed to ensure that, after sorting the positive fraction, an appropriate level of proviral DNA was present for monitoring HIV-l reactivation in the resting-like GFP negative fraction.
  • the cells were centrifuged and re-suspended in R20 to a final concentration of 5-7 M/mL in a 15 mL tube. The cells were then placed on ice until sorting.
  • the GFP negative fraction was sorted using a BD FACS Aria II Cell Sorter (BD Biosciences, San Jose, CA, USA) or a similar device.
  • the cells were cultured in a T25 flask to a final concentration of 0.8 M/mL in R10+ geneticin 0.5 mg/mL medium in the presence of 1 mM of ritonavir (RTV, Cat. No. A2169, Sigma-Aldrich Corp., Saint Louis, MO, USA). The cells were cultured during 4 days and were monitored for cell growth. Fresh media + RTV were added if the culture turned yellow.
  • RTV ritonavir
  • the cell culture was enriched in HIV-l latently infected cells (or Resting-Like“RELI” cells). Under these experimental conditions, the RELI cells had 0.6-0.9 copies of HIV-l proviral DNA per million cells.
  • RELI cells were counted and added to round-bottom 96-well plates until a final concentration of lM/mL in R10 + raltegravir (RLT 100 nM, Cat. No. CDS023737, Sigma-Aldrich Corp., Saint Louis, MO, USA) medium per well was attained.
  • Latency reactivating agents were added for 48 hours to the RELI cells to assess viral reactivation. See Table 1. LRAs were also added to uninfected cells to assess cellular toxicity. All LRA-treated cells were assessed in both co-culture and non-co-culture conditions in duplicates.
  • the HIV-l reactivation "SHOCK" was analyzed by the percentage of intracellular viral protein p24 from non-dead RELI cells.
  • CTLs were counted and their viability was checked.
  • the CTLs were seeded at a final concentration of 2 M/mL in 24-well plates with a RPMI 1640 medium containing 50% of human AB serum (Cat. No. H3667, Sigma-Aldrich Corp., Saint Louis, MO, USA).
  • the CTLs were cultured overnight in order to reduce cytokine background production.
  • the RELI cells were washed at least twice with PBS IX for 5 minutes at 800 rpm. Afterwards, the cells were re-suspended in a LRA- free R10 + RLT (100 nM) medium. RMD-treated cells were washed 5 hours post- reactivation.
  • the RELI cells/CTL co-culture was set up in round-bottom 96-well plates. CTLs were counted and re-suspended in RPMI with 10% of human AB serum (Cat. No. H3667, Sigma-Aldrich Corp., Saint Louis, MO, USA) to a final concentration of 1 M/mL. A CDl07a-PE-Cy5 antibody (clone H4A3, Cat. No. 555802, BD Biosciences, San Jose, CA, USA) was added to the CTL culture. The CTLs were then mixed with the reactivated RELI cells in a 1 : 1 ratio (100,000 RELI cells: 100,000 CTLs). The CTLs and RELI cells were co cultured for 20 hours.
  • the cells were incubated at 37°C for 3 hours in the presence of brefeldin A at 2.5 pL/mL (Golgiplug solution, Cat. No. 555029, BD Biosciences, San Jose, CA, USA) and monensin at 1.6 pL/mL (GolgiStop solution, Cat. No. 554724, BD Biosciences, San Jose, CA, USA) diluted in PBS IX (50 pL/well).
  • brefeldin A 2.5 pL/mL
  • monensin at 1.6 pL/mL
  • PBS IX 50 pL/well
  • the culture supernatant was removed.
  • the culture supernatant was removed.
  • CD8-V450 antibody pacific blue, clone RPA-T8, Cat. No. 558207, BD Biosciences, San Jose, CA, USA
  • PBS IX PBS IX
  • the cells were washed twice with 200 pL PBS IX at 800 g for 3 minutes. The culture supernatant was removed. Alternatively, the cells can be stored overnight in the Fixation Medium at 4°C. 11)
  • the following antibodies (i) p24-HIV (clone KC57- RD1, Cat. No. 6604667, Beckman Coulter, Inc., Brea, CA, USA), (ii) MIRIb- FITC (clone 24006, Cat. No. IC271F, R&D Systems, Inc. Minneapolis, MN, USA) and (iii) IFNg-PE-Cy7 (clone 4S.B3, Cat. No.
  • the CTL“KILL” was calculated taking into account the reduction of non-dead, CD8 negative, p24 positive cells from co-cultures of RELI cells and CTLs, i.e. 100- [(% p24 in non-dead CD8 negative cells co-cultured with CD8 positive cells / % p24 in non-dead CD8 negative cells in the absence of CD8 positive cells) x 100]
  • the CTL activation profile in response to viral reactivation was analyzed by the secretion of CD 107a, MIPlb and IFNg from live, CD8 positive CTLs.
  • Banerjee C, et al. BET bromodomain inhibition as a novel strategy for reactivation of HIV-l, J Leukoc Biol 2012; 92: 1147-1154.
  • HDACis Histone deacetylase inhibitors
  • Boehm D, et al. BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism, Cell Cycle 2013: 12:452-462.
  • Bosque A et al. , Induction of HIV-l latency and reactivation in primary memory CD4+ T cells, Blood 2009; 113:58-65.
  • Contreras X, et al. Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells, J Biol Chem 2009; 284:6782-6789.
  • Gallastegui E, et al. Combination of biological screening in a cellular model of viral latency and virtual screening identifies novel compounds that reactivate HIV-l, J Virol 2012; 86:3795-3808.
  • Hermankova M, et al. Analysis of human immunodeficiency virus type 1 gene expression in latently infected resting CD4+ T lymphocytes in vivo , J Virol 2003; 77:7383-7392.
  • Kauder S, et al. Epigenetic regulation of HIV- 1 latency by cytosine methylation, PLoS Pathog 2009; 5:el000495Korin Y, et al. , Effects of prostratin on T cell activation and human immunodeficiency virus latency, J Virol 2002; 76:8118-8123.
  • Keedy K, et al. A limited group of class I histone deacetylases acts to repress human immunodeficiency virus type 1 expression J Virol 2009; 83:4749-4756.
  • Rasmussen T, et al. Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial, The Lancet HIV 2014; l(l):el3-e2l.
  • Shan L, et al. Stimulation of HIV-l- specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation, Immunity 2012; 36:491- 501. 55. Shan L, et al. , A novel PCR assay for quantification of HIV-l RNA, J Virol 2013; 87(1 l):652l-6525.
  • Histone deacetylase inhibitor romidepsin induces HIV expression in CD4 T cells from patients on suppressive antiretroviral therapy at concentrations achieved by clinical dosing, PLoS Pathog 2014; l0(4):el00407l.
  • Williams S, et al. Prostratin antagonizes HIV latency by activating NF-kB, J Biol Chem 2004; 279:42008-42017.
  • Williams S, et al. NF-kB p50 promotes HIV latency through HD AC recruitment and repression of transcriptional initiation, EMBO J 2006; 25: 139-149.
  • liver is a major organ for clearing simian immunodeficiency virus in rhesus monkeys, J Virol 2002; 76(l0):527l-5273.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne un procédé avec des agents d'inversion de latence du VIH-1 (LRA). Le procédé comprend les étapes consistant à : (a) cultiver in vitro une fraction de lymphocytes T CD4+ obtenus à partir d'un sujet infecté par le VIH avec au moins un agent d'inversion de latence (LRA) pendant au moins 48 heures, (b) co-cultiver in vitro la fraction de lymphocytes T CD4+ avec des lymphocytes-T cytotoxiques (CTL) obtenus à partir du sujet infecté par le VIH et (c) compter les lymphocytes T CD4+ tués dans la fraction.
PCT/IB2018/001456 2017-11-22 2018-11-21 Procédé de criblage d'agents d'inversion de latence du vih-1 WO2019106427A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762590081P 2017-11-22 2017-11-22
US62/590,081 2017-11-22

Publications (2)

Publication Number Publication Date
WO2019106427A2 true WO2019106427A2 (fr) 2019-06-06
WO2019106427A3 WO2019106427A3 (fr) 2019-10-17

Family

ID=66379947

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2018/001456 WO2019106427A2 (fr) 2017-11-22 2018-11-21 Procédé de criblage d'agents d'inversion de latence du vih-1

Country Status (1)

Country Link
WO (1) WO2019106427A2 (fr)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016063251A1 (fr) * 2014-10-23 2016-04-28 Centre Hospitalier Universitaire Vaudois Réactifs et procédés de réactivation du vih latent

Non-Patent Citations (99)

* Cited by examiner, † Cited by third party
Title
ADLER M ET AL., BRIT. MED. J., vol. 294, 1987, pages 1145 - 1147
ARCHIN N ET AL., NATURE, vol. 487, 2012, pages 482 - 485
ARCHIN N ET AL.: "Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy", NATURE, vol. 487, 2012, pages 482 - 485, XP055560367, DOI: doi:10.1038/nature11286
ARCHIN N ET AL.: "Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid", AIDS RES HUM RETROVIRUSES, vol. 25, 2009, pages 207 - 212
ARCHIN N ET AL.: "Expression of latent human immunodeficiency type 1 is induced by novel and selective histone deacetylase inhibitors", AIDS, vol. 23, 2009, pages 1799 - 1806, XP055240700, DOI: doi:10.1097/QAD.0b013e32832ec1dc
ARCHIN N ET AL.: "HIV-1 Expression within resting CD4 T cells following multiple doses of vorinostat", J INFECT DIS, vol. 210, no. 5, 2014, pages 728 - 735
BANERJEE C ET AL.: "BET bromodomain inhibition as a novel strategy for reactivation of HIV-1", J LEUKOC BIOL, vol. 92, 2012, pages 1147 - 1154, XP055167532, DOI: doi:10.1189/jlb.0312165
BARTHOLOMEEUSEN K ET AL.: "Histone deacetylase inhibitors (HDACis) that release the positive transcription elongation factor b (P-TEFb) from its inhibitory complex also activate HIV transcription", J BIOL CHEM, vol. 288, 2013, pages 14400 - 14407
BEANS E ET AL.: "Highly potent, synthetically accessible prostratin analogs induce latent HIV expression In vitro and ex vivo", PROC NATL ACAD SCI USA, vol. 110, 2013, pages 11698 - 11703
BLAZKOVA J ET AL.: "Effect of histone deacetylase inhibitors on HIV production in latently infected, resting CD4(+) T cells from infected individuals receiving effective antiretroviral therapy", J INFECT DIS, vol. 206, 2012, pages 765 - 769, XP055431479, DOI: doi:10.1093/infdis/jis412
BOEHM D ET AL.: "BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism", CELL CYCLE, vol. 12, 2013, pages 452 - 462, XP055167537, DOI: doi:10.4161/cc.23309
BOSQUE A ET AL.: "Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells", BLOOD, vol. 113, 2009, pages 58 - 65
BOUCHAT S ET AL.: "Histone methyltransferase inhibitors induce HIV-1 recovery in resting CD4(+) T cells from HIV-1 -infected HAART-treated patients", AIDS, vol. 26, 2012, pages 1473 - 1482
BULLEN C ET AL.: "New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo", NAT MED, vol. 20, 2014, pages 425 - 429, XP055312788, DOI: doi:10.1038/nm.3489
BURNETT J ET AL.: "Combinatorial latency reactivation for HIV-1 subtypes and variants", J VIROL, vol. 84, 2010, pages 5958 - 5974
CHOMONT N ET AL., NAT MED, vol. 15, no. 8, 2009, pages 893 - 900
CHOMONT N ET AL.: "HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation", NAT MED, vol. 15, no. 8, 2009, pages 893 - 900
CHOU T: "Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies", PHARMACOL REV, vol. 58, 2006, pages 621 - 681, XP055151376, DOI: doi:10.1124/pr.58.3.10
CHOUDHARY S ET AL.: "Hexamethylbisacetamide and disruption of human immunodeficiency virus type 1 latency in CD4(+) T cells", J INFECT DIS, vol. 197, 2008, pages 1162 - 1170
CHUN T ET AL., PROC NATL ACAD SCI USA, vol. 94, 1997, pages 13193 - 13197
CHUN T ET AL.: "In vivo fate of HIV- 1 -infected T cells: quantitative analysis of the transition to stable latency", NAT MED, vol. 1, 1995, pages 1284 - 1290, XP008037535, DOI: doi:10.1038/nm1295-1284
CHUN T ET AL.: "Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy", PROC NATL ACAD SCI USA, vol. 94, 1997, pages 13193 - 13197, XP002973106, DOI: doi:10.1073/pnas.94.24.13193
CHUN T ET AL.: "Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection", NATURE, vol. 387, 1997, pages 183 - 188, XP002150582, DOI: doi:10.1038/387183a0
CILLO A ET AL.: "Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy", PROC NATL ACAD SCI USA, vol. 96, no. 26, 2014, pages 15109 - 15114
CLUTTON G ET AL., SCI REP, vol. 6, 2016, pages 30749
CLUTTON G ET AL.: "The differential short- and long-term effects of HIV-1 latency-reversing agents on T cell function", SCI REP, vol. 6, 2016, pages 30749
CONTRERAS X ET AL.: "Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells", J BIOL CHEM, vol. 284, 2009, pages 6782 - 6789
DAR R ET AL.: "Screening for noise in gene expression identifies drug synergies", SCIENCE, vol. 344, 2014, pages 1392 - 1396
DECHRISTOPHER B ET AL.: "Designed, synthetically accessible bryostatin analogues potently induce activation of latent HIV reservoirs in vitro", NAT CHEM, vol. 4, 2012, pages 705 - 710
DEEKS S ET AL., NATURE, vol. 487, 2012, pages 439 - 440
DEEKS S ET AL.: "HIV: Shock and kill", NATURE, vol. 487, 2012, pages 439 - 440
DELAGREVERIE H ET AL.: "Ongoing clinical trials of human immunodeficiency virus latency-reversing and immunomodulatory agents", OPEN FOR INFECT DIS, 2016
DESCOURS B ET AL., CLIN INFECT DIS, vol. 54, no. 10, 2012, pages 1495 - 503
DESCOURS B: "Immune responses driven by protective human leukocyte antigen alleles from long-term nonprogressors are associated with low HIV reservoir in central memory CD4 T cells", CLIN INFECT DIS, vol. 54, no. 10, 2012, pages 1495 - 503
DOYON G ET AL.: "Disulfiram reactivates latent HIV-1 expression through depletion of the phosphatase and tensin homolog", AIDS, vol. 27, no. 2, 2013, pages F7 - F11
ELLIOTT J ET AL.: "Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy", PLOS PATHOG, vol. 10, 2014, pages el004473
FINZI D ET AL., NAT MED, vol. 5, 1999, pages 512 - 517
FINZI D ET AL., SCIENCE, vol. 278, 1997, pages 1295 - 1300
FINZI D ET AL.: "Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy", SCIENCE, vol. 278, 1997, pages 1295 - 1300, XP002972795, DOI: doi:10.1126/science.278.5341.1295
FINZI D ET AL.: "Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy", NAT MED, vol. 5, 1999, pages 512 - 517
FRIEDMAN J ET AL.: "Epigenetic silencing of HIV-1 by the histone H3 lysine 27 methyltransferase enhancer of Zeste 2", J VIROL, vol. 85, 2011, pages 9078 - 9089, XP002691445, DOI: doi:10.1128/JVI.00836-11
GALLASTEGUI E ET AL.: "Combination of biological screening in a cellular model of viral latency and virtual screening identifies novel compounds that reactivate HIV-1", J VIROL, vol. 86, 2012, pages 3795 - 3808, XP055355893, DOI: doi:10.1128/JVI.05972-11
HERMANKOVA M ET AL., J VIROL, vol. 77, 2003, pages 7383 - 7392
HERMANKOVA M ET AL.: "Analysis of human immunodeficiency virus type 1 gene expression in latently infected resting CD4+ T lymphocytes in vivo", J VIROL, vol. 77, 2003, pages 7383 - 7392
HILL A ET AL.: "Predicting the outcomes of treatment to eradicate the latent reservoir for HIV-1", PROC NATL ACAD SCI USA, vol. 111, no. 37, 2014, pages 13475 - 13480
HO Y ET AL.: "Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure", CELL, vol. 155, 2013, pages 540 - 551, XP028757996, DOI: doi:10.1016/j.cell.2013.09.020
JONES R ET AL., PLOSPATHOG, vol. 10, 2014, pages el004287
JONES R ET AL.: "Histone deacetylase inhibitors impair the elimination of HIV-infected cells by cytotoxic T-lymphocytes", PLOSPATHOG, vol. 10, 2014, pages el004287
KAUDER S ET AL.: "Epigenetic regulation of HIV- 1 latency by cytosine methylation", PLOS PATHOG, vol. 5, 2009, pages el000495
KEEDY K ET AL.: "A limited group of class I histone deacetylases acts to repress human immunodeficiency virus type 1 expression", J VIROL, vol. 83, 2009, pages 4749 - 4756, XP055240698, DOI: doi:10.1128/JVI.02585-08
KLATT N ET AL.: "CD8+ lymphocytes control viral replication in SIVmac239-infected rhesus macaques without decreasing the lifespan of productively infected cells", PLOS PATHOG, vol. 6, no. 1, 2010, pages e1000747
KLOVERPIIS H ET AL., J VIROL, vol. 87, no. 19, 2013, pages 10889 - 10894
KLOVERPRIS H ET AL.: "HLA-specific intracellular epitope processing shapes an immunodominance pattern for HLA-B*57 that is distinct from HLA-B*58:01", J VIROL, vol. 87, no. 19, 2013, pages 10889 - 10894
KORIN Y ET AL.: "Effects of prostratin on T cell activation and human immunodeficiency virus latency", J VIROL, vol. 76, 2002, pages 8118 - 8123, XP055016775, DOI: doi:10.1128/JVI.76.16.8118-8123.2002
LAIRD G ET AL.: "Rapid quantification of the latent reservoir for HIV-1 using a viral outgrowth assay", PLOS PATHOG, vol. 9, 2013, pages el003398
MARKOWITZ M ET AL.: "A novel antiviral intervention results in more accurate assessment of human immunodeficiency virus type 1 replication dynamics and T cell decay in vivo", J VIROL, vol. 77, no. 8, 2003, pages 5037 - 5038
MBONYE U ET AL.: "Transcriptional control of HIV latency: cellular signaling pathways, epigenetics, happenstance and the hope for a cure", VIROLOGY, vol. 454-455, 2014, pages 328 - 339
NOEL N ET AL., J VIROL, vol. 90, no. 13, 2016, pages 6148 - 6158
NOEL N ET AL.: "Long-term spontaneous control of HIV-1 is related to low frequency of infected cells and inefficient viral reactivation", J VIROL, vol. 90, no. 13, 2016, pages 6148 - 6158
NOVIS C ET AL.: "Reactivation of latent HIV-1 in central memory CD4(+) T cells through TLR-1/2 stimulation", RETROVIROLOGY, vol. 10, 2013, pages 119, XP021166673, DOI: doi:10.1186/1742-4690-10-119
PAYNE R ET AL., J VIROL, vol. 84, no. 20, 2010, pages 10543 - 10557
PAYNE R ET AL.: "Efficacious early antiviral activity of HIV Gag- and Pol-specific HLA-B 2705-restricted CD8+ T cells", J VIROL, vol. 84, no. 20, 2010, pages 10543 - 10557
PERELSON A ET AL.: "HIV-1 dynamics In vivo: virion clearance rate, infected cell lifespan, and viral generation time", SCIENCE, vol. 271, no. 5255, 1996, pages 1582 - 1586
PRADO J ET AL., J VIROL, vol. 84, no. 1, 2010, pages 492 - 502
PRADO J ET AL.: "Replicative capacity of human immunodeficiency virus type 1 transmitted from mother to child is associated with pediatric disease progression rate", J VIROL, vol. 84, no. 1, 2010, pages 492 - 502
PRINS J ET AL., AIDS, vol. 13, 1999, pages 2405 - 2410
PRINS J ET AL.: "Immuno-activation with anti-CD3 and recombinant human JL-2 in HIV- 1 -infected patients on potent antiretroviral therapy", AIDS, vol. 13, 1999, pages 2405 - 2410
RASMUSSEN T ET AL.: "Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial", THE LANCET HIV, vol. 1, no. 1, 2014, pages e13 - e21
REUSE S ET AL.: "Synergistic activation of HIV-1 expression by deacetylase inhibitors and prostratin: implications for treatment of latent infection", PLOS ONE, vol. 4, 2009, pages e6093
SACHA J ET AL., NAT PROTOC, vol. 5, no. 2, 2010, pages 239 - 246
SACHA J ET AL.: "Synchronous infection of SIV and HIV in vitro for virology, immunology and vaccine-related studies", NAT PROTOC, vol. 5, no. 2, 2010, pages 239 - 246, XP055419218, DOI: doi:10.1038/nprot.2009.227
SHAN L ET AL., IMMUNITY, vol. 36, no. 3, 2012, pages 491 - 501
SHAN L ET AL.: "A novel PCR assay for quantification of HIV-1 RNA", J VIROL, vol. 87, no. 11, 2013, pages 6521 - 6525, XP055488620, DOI: doi:10.1128/JVI.00006-13
SHAN L ET AL.: "Stimulation of HIV-1- specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation", IMMUNITY, vol. 36, 2012, pages 491 - 501, XP028475174, DOI: doi:10.1016/j.immuni.2012.01.014
SHAN L ET AL.: "Unique characteristics of histone deacetylase inhibitors in reactivation of latent HIV-1 in Bcl-2-transduced primary resting CD4+ T cells", ANTIMICROB CHEMOTHER, vol. 69, no. 1, 2014, pages 28 - 33, XP055240694, DOI: doi:10.1093/jac/dkt338
SILICIANO J ET AL., NAT MED, vol. 9, 2003, pages 727 - 728
SILICIANO J ET AL.: "Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells", NAT MED, vol. 9, 2003, pages 727 - 728
SINGH A ET AL.: "Noise in viral gene expression as molecular switch for viral latency", CURR OPIN MICROBIOL, vol. 12, no. 4, 2009, pages 460 - 466, XP026439348
SMITH B ET AL.: "Differentiation therapy in poor risk myeloid malignancies: results of a dose finding study of the combination biyostatin-1 and GM-CSF", LEUK RES, vol. 35, 2011, pages 87 - 94, XP027561475
SOGAARD O ET AL.: "The HDAC inhibitor romidepsin is safe and effectively reverses HIV-1 latency in vivo as measured by standard clinical assays", INTERNATIONAL AIDS SOCIETY 20TH INTERNATIONAL AIDS CONFERENCE ON, 20 July 2014 (2014-07-20)
SPINA C ET AL.: "An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients", PLOS PATHOG, vol. 9, no. 12, 2013, pages el003834
SPIVAK A ET AL.: "A pilot study assessing the safety and latency-reversing activity of disulfiram in HIV-1-infected adults on antiretroviral therapy", CLIN INFECT DIS, vol. 58, 2014, pages 883 - 890
STRAIN M ET AL., PROC NATL ACAD SCI USA, vol. 100, 2003, pages 4819 - 4824
STRAIN M ET AL.: "Heterogeneous clearance rates of long-lived lymphocytes infected with HIV: intrinsic stability predicts lifelong persistence", PROC NATL ACAD SCI USA, vol. 100, 2003, pages 4819 - 4824
UNAIDS, November 2017 (2017-11-01), Retrieved from the Internet <URL:http://www.unaids.ofg>
WALTER-SPERLING V ET AL.: "The effect of latency reversal agents on primary CD8+ T cells: implications for shock and kill strategies for human immunodeficiency virus eradication", EBIOMEDICINE, vol. 8, 2016, pages 217 - 229
WALTER-SPERLING V, EBIOMEDICINE, vol. 8, 2016, pages 217 - 229
WEI D ET AL., PLOS PATHOG, vol. 10, no. 4, 2014, pages e1004071
WEI D ET AL.: "Histone deacetylase inhibitor romidepsin induces HIV expression in CD4 T cells from patients on suppressive antiretroviral therapy at concentrations achieved by clinical dosing", PLOS PATHOG, vol. 10, no. 4, 2014, pages e1004071, XP055240697, DOI: doi:10.1371/journal.ppat.1004071
WILLIAMS S ET AL.: "NF-kB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation", EMBO J, vol. 25, 2006, pages 139 - 149
WILLIAMS S ET AL.: "Prostratin antagonizes HIV latency by activating NF-kB", J BIOL CHEM, vol. 279, 2004, pages 42008 - 42017, XP002380467, DOI: doi:10.1074/jbc.M402124200
WONG J ET AL.: "In vivo CD8+ T cell suppression of siv viremia is not mediated by CTL clearance of productively infected cells", PLOS PATHOG, vol. 6, no. 1, 2010, pages e1000748
WONG J ET AL.: "Recovery of replication-competent HIV despite prolonged suppression of plasma viremia", SCIENCE, vol. 278, 1997, pages 1291 - 1295
XING S ET AL., DRUG DISCOV TODAY, vol. 18, no. 11-12, 2013, pages 541 - 551
XING S ET AL.: "Disulfiram reactivates latent HIV-1 in a Bcl-2-transduced primary CD4+ T cell model without inducing global T cell activation", J VIROL, vol. 85, 2011, pages 6060 - 6064, XP002785213, DOI: doi:10.1128/JVI.02033-10
XING S ET AL.: "Novel structurally related compounds reactivate latent HIV- 1 in a bcl-2-transduced primary CD4+ T cell model without inducing global T cell activation", J ANTIMICROB CHEMOTHER, vol. 67, 2012, pages 398 - 403, XP055367557, DOI: doi:10.1093/jac/dkr496
XING S ET AL.: "Targeting HIV latency: pharmacologic strategies toward eradication", DRUG DISCOV TODAY, vol. 18, no. 11-12, 2013, pages 541 - 551, XP055535176, DOI: doi:10.1016/j.drudis.2012.12.008
YANG H ET AL.: "Small-molecule screening using a human primary cell model of HIV latency identifies compounds that reverse latency without cellular activation", J CLIN INVEST, vol. 119, 2009, pages 3473 - 3486, XP009159909, DOI: doi:10.1172/JCI39199
ZHANG L: "The liver is a major organ for clearing simian immunodeficiency virus in rhesus monkeys", J VIROL, vol. 76, no. 10, 2002, pages 5271 - 5273

Also Published As

Publication number Publication date
WO2019106427A3 (fr) 2019-10-17

Similar Documents

Publication Publication Date Title
Abner et al. HIV “shock and kill” therapy: In need of revision
Gougeon Apoptosis as an HIV strategy to escape immune attack
Schlaepfer et al. TLR7/8 triggering exerts opposing effects in acute versus latent HIV infection
Andersen et al. Natural product-derived compounds in HIV suppression, remission, and eradication strategies
US20160095850A1 (en) Methods and compositions for treatment of hiv infection
US9943536B2 (en) Reactivation of HIV-1 gene expression to treat persistent HIV infection
Gougeon To kill or be killed: how HIV exhausts the immune system.
WO2016196471A1 (fr) Procédés et compositions pour le traitement d&#39;une infection par le vih
Desimio et al. The histone deacetylase inhibitor SAHA simultaneously reactivates HIV-1 from latency and up-regulates NKG2D ligands sensitizing for natural killer cell cytotoxicity
Yang et al. Chidamide, a histone deacetylase inhibitor-based anticancer drug, effectively reactivates latent HIV-1 provirus
US20200163924A1 (en) Compositions and methods for reactivating latent viral infections
US20230087766A1 (en) Methods of reactivating latent human immunodeficiency virus and related compositions
Massanella et al. Attacking the HIV reservoir from the immune and viral perspective
US20150099012A1 (en) Methods for activating retrovirus in latent infected cells, and compounds for use therein
WO2016063251A1 (fr) Réactifs et procédés de réactivation du vih latent
Boelaert et al. Chloroquine exerts an additive in vitro anti-HIV type 1 effect when associated with didanosine and hydroxyurea
US20150031739A1 (en) Panobinostat for use in the treatment of hiv-1
BRPI0513321B1 (pt) Vírus parapox em combinação com outros agentes antivirais para o tratamento de doenças virais
Katlama et al. Barriers to a cure: new concepts in targeting and eradicating HIV-1 reservoirs
Garvey et al. Response to Mallet et al.,‘Nodular regenerative hyperplasia is a new cause of chronic liver disease in HIV-infected patients’
US20180169108A1 (en) Drug Combinations For the Treatment of HIV
WO2019106427A2 (fr) Procédé de criblage d&#39;agents d&#39;inversion de latence du vih-1
Zhou et al. Bcl-2 Antagonist Obatoclax Reactivates Latent HIV-1 via the NF-κB Pathway and Induces Latent Reservoir Cell Apoptosis in Latently Infected Cells
WO2005000405A2 (fr) Compositions comprenant des inhibiteurs de la kinase p38 et leurs methodes d&#39;utilisation
Sluis-Cremer Therapeutic approaches to eradicate latent HIV-1 in resting CD4+ T Cells

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18874988

Country of ref document: EP

Kind code of ref document: A2