curing hiv: coping with hope -...

41
Curing HIV: Coping with Hope Ronald Mitsuyasu, MD Professor of Medicine UCLA Center for Clinical AIDS Research and Education (CARE Center)

Upload: others

Post on 24-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Curing HIV:

    Coping with Hope

    Ronald Mitsuyasu, MD Professor of Medicine

    UCLA Center for Clinical AIDS Research

    and Education (CARE Center)

  • Curing HIV is:

    0%

    0%

    0%

    0%

    0%

    0% A. Impossible

    B. Requires removing all HIV from the host

    C. Means controlling HIV without antiretroviral therapy

    D.Requires high dose chemotherapy

    E. Is only possible in newborns

    F. B + D

  • Possible approaches to “curing” HIV are:

    0%

    0%

    0%

    0%

    0%A. Continuing ART for > 70 yrs with no missed doses

    B. Requires inducing HIV from latent reservoirs

    C. Only occurs with bone marrow or stem cell transplants

    D. Will likely require multiple strategies

    E. Means the person cannot be infected again with HIV

  • Antiretroviral Drugs 2014 (28)

    Nucleosidetide analogues • zidovudine (zdv)

    • didanosine (ddI)

    • zalcitabine (ddC)

    • stavudine (d4T)

    • lamivudine (3TC)

    • abacavir (ABC)

    • emtricitabine (FTC)

    • tenofovir (TFV)

    Non-nucleoside analogues

    • nevirapine (NVP)

    • delavirdine (DLV)

    • efavirenz (EFV)

    • etravirine (ETV)

    • rilpivirine (RPV)

    Protease Inhibitors (10)

    • saquinavir (SQV)

    • ritonavir (RTV)

    • indinavir (IDV)

    • nelfinavir (NFV)

    • amprenavir (APV)

    • lopinavir/r (LPV/r)

    • fosamprenavir (FPV)

    • atazanavir (ATV)

    • tipranavir (TPV)

    • darunavir (DRV)

    Reverse Transcriptase Inhibitors(13)

    Integrase Inhibitor (3) •raltegravir (RAL)

    •elvitegravir (ELV)

    •dolutegravir (DTG)

    Fusion Inhibitor •Infuvirtide (Fuzeon)

    Entry Inhibitor (CCR5) •maraviroc (MVC)

  • Problems with Current ARV Therapies

    Must be taken continuously

    Does not eradicate HIV. Only inhibits replication

    There are side effects and toxicities with each class of ARV drugs

    Difficulty with long term adherence (fatigue factor)

    Resistance development

    Expense and inconvenience of chronic therapy

  • Major Problem with ART

    Federal Resources / Policies/Issues : HIV/AIDS Care Continuum

    http://aids.gov/federal-resources/index.html

  • What Do We Mean By “Cure”?

    Sterilizing Cure

    • Eradication of all HIV from the individual

    • No HIV found in any tissue (blood, BM, LN, GI, CSF)

    • Cannot detect HIV by most sensitive assays

    • No recurrence of HIV upon treatment interruption

    Functional Cure

    • No measurable HIV RNA in blood after long term

    treatment interruptionn

    • Maintain stable immune function off ART

    • May or may not be virus free

  • Functional Cure of the “Berlin Patient”

    with chemotherapy and HSCT

    Hutter G, et al NEJM 2009,360:692-698.

    Hutter G and Thiel. AIDS 2011,25:273-4.

  • Possible Reasons for Non-detectable HIV off ART

    (remission) in the “Berlin Patient”

    Long term ART had reduced HIV burden

    Chemotherapy removed infected cells in patient

    Transplanted cells protected from HIV infection

    due to CCR5 delta 32 mutation

    Unrelated donor cells contributed to a GVH-like

    reaction further clearance of latently infected cells

    Generation of host protective immunity

    Combination of above

    Did they just get lucky?

  • Eradication Not So Simple

    Latently-infected resting memory CD4+ T

    cells persist as viral reservoirs despite

    years of ART

    Virus RNA detected in blood and other

    lymphoid tissues of patients suppressed

    for >10 years

    t1/2 of latently-infected resting memory

    CD4+ T cells is ~ 44 months making their

    elimination possible in ~70 years (if fully

    compliant with no breakthrough)

  • Interventions to possibly eliminate

    long-lived HIV-infected cells

    Block ongoing, low level HIV replication

    • ART, anti-microbial, anti-inflammatory, anti-coagulants, antifibrotics, anti-aging drugs

    Activate HIV-1 expression from latently-infected CD4+ memory T-cells, “Kick and Kill”

    • Assumes this is the key reservoir

    • Assumes activation is lethal for latently-infected cell

    Selectively kill cells expressing HIV-1

    • Generate antibodies or CI to HIV infected cells

    Non-selectively kill all or most cells which harbor HIV with chemo-RT and allow host immune system to mop up any residual reservoir cells (may require further immune activation or HIV vaccination)

    Protect new cells from HIV infection

  • Starting ART VERY Early

    Mississippi baby: started ART within 31 hours;

    stopped ART at 18 mos; now thought cured at age 41

    mos (23 months off ART)

    • Undetectable RNA; Trace HIV-1 DNA detectable

    Persaud NEJM 2013; 369; 1828

    Long Beach baby: started ART within 4 hours; +HIV

    DNA PCR (4 hrs) and +HIV RNA (36 hrs); +CSF HIV

    PCR (day 6); no HIV RNA; no proviral HIV DNA after

    6 days; now age 9 months on ART Persaud CROI 2014 #75LB

  • HIV-1 Reservoirs Reduced in HIV-Positive

    Children With Early ART and Viral Control

    144 perinatally HIV-infected

    pts with long-term (median:

    10.2 yrs) virologic

    suppression on ART

    Higher proviral burden with

    increasing age at virologic

    suppression

    In perinatally infected baby

    treated early (at 4 hrs of age)

    with triple ART, noninduced

    proviral genomes detected by

    PCR at 1 mo but not at 3 mos

    of age

    Persaud D, et al. CROI 2014. Abstract 72.

    Persaud D, et al. CROI 2013. Abstract 48LB.

    Proviral Reservoir Size by Age of

    Virologic Control

    Age, yr Median copies HIV DNA/ 106 PBMCs (IQR)

    < 1

    (n = 14)

    4.2 (2.6-8.6)

    1-5

    (n = 53)

    19.4 (5.5-99.8)

    > 5

    (n =77)

    70.7 (23.2-70.7)*

    *P < .001 compared with < 1 yr

  • Starting ART VERY Early

    Patient on PrEP Demonstration Project

    HIV- at screening but HIV+ HIV at baseline

    RNA 220 cps/ml

    TDF/FTC X 7d (RNA 120), then cART (

  • “Kick and Kill” Strategies

    Epigenetic inducers/inhibitors

    • HDAC inhibitors: Voriniostat, romidepsin

    • Proteosome inhibitors: Bortezomib

    • DNA methylation inhibitors: Azacytidine, decitabine

    • HAT and DNMT inhibitor: Flavonoids, hydralazine

    • oxazolines, isoxazolines

    Non-specific activators

    • Prostratin, Valproate, Disulfiram

    • Protein kinase C activators: Bryostatin (+/-

    nanoparticles)

    Immune induction/activation

    • IL-2, IL-7, IL-11

    • Anti-CD3 antibodies (OKT3)

    • Immune-toxin conjugates

  • Multiple doses of vorinostat

    Margolis CROI 2014, # 435 LB

  • Chemokine receptor

    inhibitors: • maraviroc, TB-652

    Anti-infective therapy: • CMV, EBV, HSV, HCV/HBV

    Microbial translocation: • sevelamer, colostrum, rifaximin

    Enhance T cell renewal: • Growth Hormone, IL-7

    Anti-fibrotic drugs: • pirfenidone, ACEi, ARBs, KGF

    Anti-aging: • caloric restriction, vit. D, omega-3

    fatty acids, rapamycin, diet,

    exercise

    • Anti-inflammatory drugs: • Chloroquine, Hydroxychloroquine

    • Minocycline

    • NSAIDs (COX-2i, aspirin)

    • Statins

    • Methotrexate

    • Anakinra (IL-1Ra)

    • Thalidomide, lenalidomide,

    pentoxyfylline

    • Biologics (TNFi, IL-6i, anti-IFNa,

    anti-PD1, anti-PDL1, JAKi, IDOi,

    Casp-1i)

    • Anti-coagulants:

    • warfarin, dabigatran, aspirin,

    clopidogrel

    • Immune enhancers:

    • HIV vaccines and HIV antibodies

    Therapeutic Interventions in Development

    Combination therapy may be necessary

  • Reduced HIV Reservoir after Allogeneic Stem Cell Transplant

    Henrich T, et al. 7th IAS Conference. Kuala Lumpur, 2013. Abstract WeLBA05 .

    .

    Patient A: PBMC DNA CD4 Count

    Post HSCT (days)

    0 200 400 600 800 1000 1200 1400 1600

    HIV

    DN

    A

    (co

    pie

    s/1

    06 P

    BM

    C)

    Post HSCT (days)

    0 200 400 600 800 1000 1200 1400 1600

    CD

    4 (

    ce

    lls

    /mm

    3) 4.3 Years

    Post-HSCT (

  • Allogeneic Stem Cell Transplant

    Patient A:

    • No HIV RNA after 15 weeks off ART

    • Rapid relapse of HIV after 32 weeks

    • Developed acute HIV symptoms

    • Genetic testing showed single/same HIV genotype

    Patient B:

    • No HIV RNA after 7 weeks off ART

    • Rapid relapse of HIV after 12 weeks

    • Developed acute HIV symptoms

    • Genetic testing showed single/same HIV genotype

    Henrich T et al CROI 2014, Boston, MA

  • Gene Therapy Definition

    Introduction of a gene

    transfer product into cells

    to produce a therapeutic

    benefit

  • Anti-HIV-1 Gene Therapy

    Recombinant T-cell receptor (CD4 zeta)

    Transdominant proteins (Rev M10, Trev, C46)

    Intracellular antibodies and RNA decoys

    Antisense (tat, RevM10, env ViRxsys)

    Ribozymes (U5 hairpin, Rz2 hammerhead)

    dsRNA (RNAi, siRNA)

    RNA aptamer (small RNA antagonists of protein function)

    Zinc finger nucleases (CCR5 directed, SB-728)

    David Baltimore, PhD – “Intracellular Immunization”

  • Anti-HIV-1 Gene Therapy

    Recombinant T-cell receptor (CD4 zeta)

    Transdominant proteins (Rev M10, Trev, C46)

    Intracellular antibodies and RNA decoys

    Antisense (tat, RevM10, env ViRxsys)

    Ribozymes (U5 hairpin, Rz2 hammerhead)

    dsRNA (RNAi, siRNA)

    RNA aptamer (small RNA antagonists of protein function)

    Zinc finger nucleases (CCR5 directed, SB-728)

    David Baltimore, PhD – “Intracellular Immunization”

  • Gene Therapy Targets

    Rossi, June, Kohn. Nat Biotech 25:1444, 2007

    Zn Finger Nuclease

    CCR5r inhibitors

    Fusion inhibitors

    Antisense Ribozymes

    TAR or RRE decoys

    shRNAs

    Aptimers

  • Rationale for Use of Gene Transfer

    Approach in HIV Infection

    Intracellular gene therapy works without medications that can effect other organ systems

    Gene therapy may have long lasting effects and can be self perpetuating if placed in appropriate stem cells

    Gene modification may inhibit HIV replication and/or protect uninfected cells

    May be effective against multiple clades and drug resistant strains of HIV

    May be less prone to development of resistance

    Progeny of a small number of gene modified cells may have a survival advantage and persists for long time

  • Challenges in Gene Therapy

    Technical Challenges

    Selecting gene target(s) and inhibitory mechanisms

    Getting good transduction efficiency

    Getting good engraftment of transduced cells

    • Conditioning regimen or transduction approach

    Apheresis and large cell processing/storage/shipping

    Monitoring cell marking and trafficking

    “Scaleability” and cost

    Subject Challenges

    Selecting appropriate patient population

    Identifying and recruiting subjects

    Long term safety follow up

  • Preliminary Results from 220 HIV+ treated

    with gene therapy

    HSC and T cells successfully harvested and gene

    modified

    In absence of conditioning, gene marking ranges

    from 0.01% to 0.38% (May need only 10%)

    No toxic effects or major side effects seen from

    procedure

    Encouraging early results for VL and CD4+ Tcell

    counts and decrease in inflammatory markers

    Selective advantage of gene modified cells

    demonstrated in blood, bone marrow and GALT

    Myeloablation or conditioning allows greater level of

    engraftment and gene marking (DiGusto et al, 2010)

    Cavazzana-Calvo, CROI 2013, Atlanta, abst 124

  • Zinc Finger Nuclease Studies

  • ZFP

    ZFP

    Zinc Finger Nucleases (ZFNs) “Designer Restriction Enzyme”

    Heterodimer comprised of 2 domains

    - Nuclease domain of FokI restriction enzyme - Zinc finger protein provides DNA binding specificity; targets 12 nucleotide each

    ZFN cleavage results in a double stranded DNA break - Non-homologous end-joining repair leads to permanent CCR5 gene modification

    Delivered with a non-integrating, replication-deficient, chimeric

    adenoviral 5/35 vector

    Approximately 16-39% (mean = 23%) of the CCR5 modifications

    contain a CTGAT duplication (Pentamer)

    DNA

    CCR5

    ZFN modification

    Site 165

    D32 mutation

  • SB-728-T GMP Manufacturing Process:

    Autologous ZFN CCR5-Disrupted CD4+ T-cells

    Leukapheresis

    Enriched

    CD4+

    Adenoviral SB-728

    transduction and

    CCR5 disruption

    of CD4 cells

    Cryopreserve

    cell product

    (SB-728-T)

    ~25% CCR5

    modification

    Infuse

    Activate with

    Anti-CD3/28

    beads

    Monocyte and

    CD8+ T cell

    depletion

    Expansion in WAVE

  • 30

    SB-728-T Increases CD4 T-cell Counts

    in Peripheral Blood after Infusion

    102 103 104

    201 203 302

    303304305.

    Median

    Cohort 1 Cohort 2 Cohort 3

    Days

    0 30 60 90 120 150 180 210 240 270 300 330 360

    Ch

    an

    ge

    in

    CD

    4 T

    -Ce

    ll C

    ou

    nt

    fro

    m B

    as

    eli

    ne

    (pe

    r L

    )

    -200

    0

    200

    400

    600

    800

    1000

    1200

    100 Cells

  • 31

    SB-728-0902

    Treatment Interruption – Subject 102

    Treatment Interruption

    Time

    0

    Day

    7

    Month

    3

    Month

    6

    Month

    9M

    onth

    12

    WE

    EK

    2

    WE

    EK

    4

    WE

    EK

    6

    WE

    EK

    7

    WE

    EK

    8W

    EE

    K 1

    0W

    EE

    K 1

    2W

    EE

    K 1

    4W

    EE

    K 1

    6W

    EE

    K 2

    0W

    EE

    K 2

    4W

    EE

    K 2

    8

    Lo

    g V

    iral

    Lo

    ad

    (C

    op

    ies/m

    L)

    101

    102

    103

    104

    105

    106

    Esti

    mate

    d C

    CR

    5 M

    od

    ifie

    d C

    D4 T

    -Cell

    s

    (% o

    f C

    D4

    Cells

    )

    0

    2

    4

    6

    8

    Viral Load

    Estimated CCR5 Modified CD4 T-Cells

    Mitsuyasu, ICAAC, 2010

  • Gene Modified Stem Cells

  • Neutrophil

    CD34+

    progenitor

    cell

    Myeloid stem cell

    T progenitor

    Thymocyte

    CD4+

    T cell B cell

    Megakaryoblast Erythroid

    progenitor

    Eosinophil

    progenitor

    Platelets Red blood cells

    Myelomonocytic

    progenitor

    Basophil

    progenitor

    Basophil Eosinophil

    Lymphoid stem cell

    Megakaryocyte

    B progenitor

    Macrophage

    Monocyte

    CD8+ T cell

    Hematopoiesis

  • Cal-1 Clinical Trial

    Sponsored by Calimmune, Inc.

    California Institute for Regenerative Medicine

    Conducted at

    UCLA CARE Center and Quest in SF

  • Anti-HIV Gene

    Autologous

    Hematopoietic Stem Cells

    A

    A

    Macrophages A

    CD4+ cells

    T lymphocyte development

    A

    Myeloid development

    A A A

    Intracellular immunization:

    Progressive population of immune system

    with cells protected against HIV

    Engineering Protection

    • Delivery to Hematopoietic Stem/Progenitor Cells for long-term

    protection

    • Delivery to CD4+ T Cells for short- to medium-term protection

  • Cal-1 is a self-inactivating lentiviral vector that contains two active anti-HIV agents

    •Sh5 is a short hairpin RNA against the HIV-1 co-receptor CCR5

    •C46 is a membrane-anchored C-peptide derived from the HIV-1 envelope glycoprotein gp41

    3

    7

    Investigational Product

    •Active against both R5- and X4- strains of HIV •Two points of inhibition for R5-tropic HIV-1 •Mitigates against resistance of HIV

  • Cell Processing Overview

    HIV-Infected individual

    Standard volume

    apheresis

    G-CSF

    Small volume

    apheresis

    CD34+ cell isolation with

    CliniMacs

    CD4+cell isolation with

    CliniMacs

    Infuse cells

    Isolated CD4+

    Transduction with Cal-1

    HSPCtn Ttn

    Isolated CD34+

    1. Apheresis

    2. Cell isolation 3. Lentiviral Transduction

    5. Autologous transplant of genetically modified cells

    4. Harvest of transduced CD4+ and CD34+ cells

    Busulfan to create bone marrow “space”

  • Directions for moving forward

    Improve gene product effects against HIV – multiple targets

    and potent constructs

    Better, safer transduction of cells (transduction >90%)

    Automate transduction procedure

    Improve engraftment of marked cells (conditioning)

    Multiple cell types (HSC and T cells M/M and others)

    Determine the best means of applying selection pressure

    for proliferation and differentiation of cells (off ART)

    Determine distribution and functionality of gene marked

    cells (in blood, BM, GALT, LN)

    Combine with other anti-HIV immune strategies (e.g.

    therapeutic vaccines)

    May be part of the “Functional Cure of HIV”

  • Curing HIV is:

    0%

    0%

    0%

    0%

    0%

    0% A. Impossible

    B. Requires removing all HIV from the host

    C. Means controlling HIV without antiretroviral therapy

    D.Requires high dose chemotherapy

    E. Is only possible in newborns

    F. B + D

  • Possible approaches to “curing” HIV are:

    0%

    0%

    0%

    0%

    0%A. Continuing ART for > 70 yrs with no missed doses

    B. Requires inducing HIV from latent reservoirs

    C. Only occurs with bone marrow or stem cell transplants

    D. Will likely require multiple strategies

    E. Means the person cannot be infected again with HIV

  • Thank You

    Questions and Comments