31
Clinical HIV-1 eradication studies Mathias Lichterfeld, M. D., Ph. D. Massachusetts General Hospital Harvard Medical School No disclosures

Clinical HIV-1 eradication studiesregist2.virology-education.com/2014/2nd_HIVFuture/6... · 2014. 10. 11. · • Can reactive HIV-1 in ex-vivo assays (Wei et al, Plos Path 2014)

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

  • Clinical HIV-1 eradication studies

    Mathias Lichterfeld, M. D., Ph. D. Massachusetts General Hospital

    Harvard Medical School

    No disclosures

  • HIV

    RN

    A (c

    ps/m

    L)

    50

    Years on cART 0 1

    1

    Slide courtesy of Javier Martinez-Picado/Sharon Lewin

    Plasma single copy assay

    Cell associated HIV DNA

    Infectious virus (IUPM)

    Blood Tissue Cell associated HIV DNA

    Cell associated HIV RNA

    HIV-1 persistence despite HAART

    >10 years

  • Eckstein, Immunity 2001; Swiggard, J Virol 2005; Saleh, Blood 2007; Marini, J Immunol 2008; Bosque, Blood 2009; Cameron, PNAS 2010; Lassen, PLoS One 2012

    Activated CD4+ T cell

    Resting CD4+ T cell

    cART

    Survival (long-half life)

    Homeostatic proliferation

    Latent and replicative HIV-1 infection in CD4 T cells

    Slide courtesy of Javier Martinez-Picado/Sharon Lewin

  • Clinical HIV-1 eradication strategies

    •Ex-vivo gene editing/gene therapy to reduce viral co-receptor

    expression, followed by adoptive immunotherapy of HIV-1 resistant cells

    • Bone marrow/HSC transplantation with CCR5-negative grafts

    • Combination of latency reversing agents with immune mediated

    interventions (“kick and kill”)

  • HMTi EZH2 inhibitors

    PKC activators Prostratin

    DNA methylation Inhibitors

    Didier Trono et, al. Science 2010

    Pharmacological latency-reversing agents “Shock” component

    HDACi Panobinostat Romidepsin Vorinostat

  • • Vaccines designed to induce effector T cell responses • Broadly-neutralizing antibodies, possibly labeled with

    Immunotoxins etc.

    • Activators of innate immunity • Immunoregulatory interventions (i. e. PD-1 inhibitors etc)

    Immune-mediated interventions “kill” component

  • TF

    OFF

    Bolden, Nat Rev Drug Disc 2006; Prince. Clin Canc Res 2009; Contreras, J Biol Chem 2009; Archin AIDS Res Hum Retrovir 2009; Reuse, PLoS One 2009; Burnett , J Virol 2010

    HDACi

    DNA nucleosomes

    HDACi turn HIV-1 transcription “on”

    Slide courtesy of Javier Martinez-Picado/Sharon Lewin

  • Clinical studies with HDACi

    Vorinostat (Merck) Romidepsin (Celgene/Gilead)

    Panobinostat (Novartis)

    Ex-vivo HIV-1 reactivation

    weaker strong strong

    Dose, dosing schedule and formulation

    400mg p. o. •Single-dose (Archin et al, Nature 2012)

    •three consecutive doses per week for eight weeks (Archin et al, JID 2014)

    •daily for 14 days (Elliott et al, CROI 2013)

    • 5mg/m2 i. v. single-dose infusion (Aarhus)

    • 0.5/2/5mg/m2 (ACTG)

    20mg p. o. TIW (M, W, F) QOW •Rasmussen et al, Lancet HIV 2014

    Pilot clinical trials in HAART-treated

    HIV patients

    completed (UNC, Melbourne)

    in process (ACTG, Aarhus)

    completed (Aarhus)

  • Archin et al, Nature 2012 and JID 2014

    Elliott/Lewin, CROI 2013

    • Single dose vorinostat led to a 4.8-fold increase in cell-associated HIV RNA.

    • No significant increases in HIV-1 RNA during repetitive dosing

    • No change in HIV-1 DNA

    • Multiple dose vorinostat led to a mean 2.5-fold increase in cell-associated HIV RNA in total CD4+ T cells.

    • No effect on CD4 T cell-associated HIV-1 DNA or plasma HIV-1 RNA

    Clinical studies with Vorinostat (Saha)

  • • Licensed in US for treatment of PTCL and CTCL

    • Can reactive HIV-1 in ex-vivo assays (Wei et al, Plos Path 2014)

    • Currently being tested in RCT (ACTG5315) (single dose at 0.5mg/m2, 2mg/m2, 5mg/m2)

    • In vivo evaluation in a non-randomized trial in six patients (Sogaard et al, IAS 2014)

    – Romidepsin (5 mg/m2) iv on day 0, 7, and 14 – Endpoints: Safety, cell-associated HIV RNA, plasma HIV RNA

    – Self-reported AEs: Total of 40, 36 related to RMD. Most common:

    • Nausea, vomiting, diarrhea, abdominal pain • Fatigue • All self-resolving, none with > grade 2

    Romidepsin

    http://en.wikipedia.org/wiki/File:Romidepsin_structure_(2).svg

  • Cell-associated HIV-1 RNA during treatment with Romidepsin

    Sogaard et al, IAS 2014

  • Viral load: COBAS® TaqMan® HIV-1 Test, v2.0 TMA: Qualitative NAT screening system (PROCLEIX ULTRIO Plus, Genprobe)

    Plasma HIV-1 RNA during treatment with Romidepsin

    Sogaard et al, IAS 2014

  • Cell-associated HIV-1 DNA during treatment with Romidepsin

    Sogaard et al, IAS 2014

  • Panobinostat (LBH589)

    • A hydroxamic acid pan-HDAC inhibitor • Developed by Novartis for the treatment

    of multiple myeloma (approval expected in 2014)

    • Dosed 30-60 mg TIW or TIW QOW • Inhibitory activity in the lower nM range

    against HDACs 1, 2 and 3 which appear important to maintaining HIV latency

    Huber et al 2011. J Biol Chem Keedy et al 2009. Journal of Virology Archin et al 2009. AIDS

    • Panobinostat induced HIV production in latently infected cell lines and primary T cells with high potency

    Rasmussen et al 2013, HVI

  • Overall study design

    • Blood draws – Twice at baseline – Twice every treatment cycle and once every pause week (13 times during panobinostat

    treatment) – 12 and 32 weeks after panobinostat initiation

    • Lumbar puncture and sigmoid biopsies before and during the last treatment cycle

  • Safety – adverse events

    • A total of 16 AEs presumed related to panobinostat (all CTCAE grade 1) • 10/15 patients experienced AEs presumed related to panobinostat • Fatigue the most frequent AE (experienced by 7/15 patients) • All completed full panobinostat dosing and follow-up

    Rasmussen et al, Lancet HIV 2014

  • • A highly statistically significant increase over time (repeated measurement ANOVA; P

  • HIV-1 plasma viremia during treatment with panobinostat

    Transcription mediated amplification (TMA)-based detection of HIV RNA:

    50% analytic sensitivity to detect 3.8 copies/ml

    95% analytic sensitivity to detect 12 copies/ml

    • Only 1/15 remained undetectable at all time points during panobinostat treatment • 4/15 patients positive at all time points (make up 8/9 positive baseline values) • 9/15 negative at both baseline samples and became positive during panobinostat treatment

    Rasmussen et al, Lancet HIV 2014

  • Total cell-associated HIV-1 DNA during treatment with panobinostat

    • Decrease from baseline to day 14 (end of first treatment cycle) • Overall no decline in total HIV DNA from baseline to week 12 (4 weeks post treatment)

    Measured per 106 CD4+ T cells using ddPCR

    Rasmussen et al, Lancet HIV 2014

  • • 4/15 patients displayed sustained reductions in total HIV DNA • No difference in nadir CD4, baseline CD4 or baseline HIV DNA

    Total cell-associated HIV-1 DNA during treatment with panobinostat

  • Total HIV DNA during panobinostat treatment – post-hoc analysis

  • Change in HIV-1 DNA during treatment with panobinostat correlates with kinetics

    of viral rebound during ATI

    • 9/15 patients agreed to participate in ATI, including three “responder patients”

    • No association between baseline characteristics and viral rebound during ATI

    Rasmussen et al, Lancet HIV 2014

    0 20 40 60 80101

    102

    103

    104

    105

    106

    107

    time after treatment interruption (days)

    plas

    ma

    viral

    load

    (cop

    ies/

    ml)

    0 20 40 60 80101

    102

    103

    104

    105

    106

    107

    time after treatment interruption (days)

    plas

    ma

    viral

    load

    (cop

    ies/

    ml)

  • Expansion of HIV-1-specific CTL during Panobinostat treatment

    0

    500

    1000

    1500

    2000

    tota

    l mag

    nitu

    de o

    fH

    IV-1

    -spe

    cific

    CTL

    (SFC

    /mill

    ion

    PBM

    C)

    p=0.007

    Magnitude Breadth

    0

    2

    4

    6

    8

    tota

    l bre

    adth

    of

    HIV

    -1-s

    peci

    fic C

    TL(n

    o of

    res

    pons

    es)

    p=0.01

    before treatment

    before treatment

    after treatment

    after treatment

    • Screening for CTL with library of optimal CTL epitopic peptides (IFN-γ Elispot)

  • HIV-1-specific CTL are not associated with HIV-1 DNA changes during

    panobinostat treatment

    Interferon-γ TNF-α

    FC HIV-1 DNA (log10) FC HIV-1 DNA (log10) FC H

    IV-1

    -spe

    cific

    effe

    ctor

    CTL

    FC H

    IV-1

    -spe

    cific

    effe

    ctor

    CTL

    • no associations between CTL magnitude or breadth and HIV-1 DNA levels • also no associations between protective HLA class I alleles and HIV-1 DNA levels

  • Expression patterns of Interferon-stimulated genes correlate with HIV-1 DNA decrease during treatment

    with panobinostat

  • HIV-1 DNA decrease during panobinostat treatment occurs preferentially in carriers of IL28B “CC”

    carriers

    CC CT0.1

    1

    10Fo

    ldc h

    ange

    into

    talH

    I V-1

    DN

    A p=0.04

    CC CT0.1

    1

    10Fo

    ldc h

    ange

    into

    talH

    IV- 1

    DN

    A p=0.04

    IL-28B

  • Changes in HIV-1 DNA during panobinostat treatment are correlated to changes in NK cells

    0.1 1 100

    1

    2

    3

    FC in HIV-1 DNA (log10)

    FC in

    CD

    69+

    NK

    cel

    ls

    0.1 1 100.0

    0.5

    1.0

    1.5

    2.0

    2.5

    FC in HIV-1 DNA (log10)

    FC in

    CD

    69+

    NK

    cel

    ls

    0.25 1 4 160

    1

    2

    3

    FC in HIV-1 DNA (log10)

    FC in

    CD

    69+

    NK

    cel

    ls

    rho:-0.64, p=0.02rho:-0.66; p=0.02 rho:-0.56; p=0.05

    Week 2 Week 6 Week 8(end of study)

    0.1 1 100

    1

    2

    3

    FC in HIV-1 DNA (log10)

    FC in

    CD

    69+

    NK

    cel

    ls

    0.1 1 100.0

    0.5

    1.0

    1.5

    2.0

    2.5

    FC in HIV-1 DNA (log10)

    FC in

    CD

    69+

    NK

    cel

    ls

    0.25 1 4 160

    1

    2

    3

    FC in HIV-1 DNA (log10)

    FC in

    CD

    69+

    NK

    cel

    ls

    rho:-0.64, p=0.02rho:-0.66; p=0.02 rho:-0.56; p=0.05

    Week 2 Week 6 Week 8(end of study)

  • Changes in HIV-1 DNA during panobinostat treatment are correlated with changes in NK cells

  • pre-IFN post-IFN100

    101

    102

    103

    104

    105HI

    V-1

    DNA

    copi

    es/1

    06 C

    D4+

    T c

    ells

    p=0.0003

    Changes in HIV-1 DNA during treatment with IFN-a/RBV in HIV/HCV co-infected patients

  • Conclusions

    • HDACi can increase HIV-1 transcription in CD4 T cells from ART-treated

    patients

    • Viral reactivation with RMD and PBT leads to transient increases of plasma RNA

    • All HDACi have an acceptable safety profile in ART-treated patients

    • No significant changes in HIV-1 DNA during HDACi treatment on population

    level

    • Some patients during treatment with PBT and RMA have substantial decline of

    HIV-1 DNA

    • Patients with reduction of HIV-1 DNA during PBT treatment differ in • IL-28B “CC” GT

    • ISG expression patterns

    • Innate effector cell activity

    • Kinetics of viral rebound during ATI

  • Department of Infectious Diseases, Aarhus University Hospital •Thomas Rasmussen, MD •Ole Schmeltz Søgaard, MD, PhD •Martin Tolstrup, MSc, PhD •Lars Østergaard, Professor/Head, MD, DMSc, PhD •Christel Rothe Brinkmann, MSc, PhD •Rikke Olesen, MD, PhD •Anni Winckelmann and Ann-Sofie Kjer •Lene Svinth Jøhnke and Erik Hagen Nielsen

    Department of Infectious Diseases, Alfred Hospital, Melbourne •Sharon Lewin, Professor/Head, PhD •Ajantha Solomon, BSc

    Massachusetts General Hospital, Ragon Institute, Boston •Maria Buzon, MSc, PhD •Selena Vigano, PhD

    Westmead Millennium Institute for Medical Research, Sydney •Sarah Palmer, PhD

    University of Colorado, School of Medicine •Charles A. Dinarello, Professor of Medicine and Immunology

    Acknowledgments

    Clinical HIV-1 eradication �studies��Mathias Lichterfeld, M. D., Ph. D.�Massachusetts General Hospital�Harvard Medical School�� �Dianummer 2Dianummer 3Clinical HIV-1 eradication strategiesPharmacological latency-reversing agents�“Shock” componentImmune-mediated interventions�“kill” componentDianummer 7Clinical studies with HDACiClinical studies with Vorinostat (Saha)Dianummer 10Dianummer 11Dianummer 12Dianummer 13Panobinostat (LBH589)Overall study designSafety – adverse eventsCell-associated unspliced HIV RNA (CA US HIV-RNA)HIV-1 plasma viremia during treatment with panobinostatTotal cell-associated HIV-1 DNA during �treatment with panobinostatTotal cell-associated HIV-1 DNA during �treatment with panobinostatTotal HIV DNA during panobinostat treatment – post-hoc analysisChange in HIV-1 DNA during treatment with panobinostat correlates with kinetics �of viral rebound during ATIExpansion of HIV-1-specific CTL �during Panobinostat treatmentHIV-1-specific CTL �are not associated with HIV-1 DNA changes during �panobinostat treatmentExpression patterns of Interferon-stimulated genes�correlate with HIV-1 DNA decrease during treatment with panobinostatHIV-1 DNA decrease during panobinostat treatment occurs preferentially in carriers of IL28B “CC” carriersDianummer 27Dianummer 28Dianummer 29ConclusionsDianummer 31