1
Evidence for an Actual Cure of a SHIV-infected Monkey Abstract Background Results José M. Martinez-Navio 1* , Sebastian P. Fuchs 1,2* , Shara N. Pantry 1 , Natasha N. Duggan 3 , William A. Lauer 1 , Eva G. Rakasz 4 , Brandon F. Keele 5 , Michel C. Nussenzweig 6,7 , Jeffrey D. Lifson 5 , Guangping Gao 8 & Ronald C. Desrosiers 1 Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. 2 Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054 Germany. 3 Department of Molecular Cell and Developmental Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. 4 Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 53711, USA. 5 AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, USA. 6 Laboratory of Molecular Immunology and 7 Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA. 8 Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, 01655, USA. *These authors contributed equally to this work. °Mentor References 3BNC117 0 6 12 18 24 30 36 0 50 100 150 200 250 Weeks post-AAV inoculation Antibody µg/mL rh2434 rh2436 rh2438 rh2443 anti-3BNC117 0 6 12 18 24 30 36 0.0 0.5 1.0 1.5 2.0 Weeks post-AAV inoculation Absorbance at 450 nm rh2434 rh2436 rh2438 rh2443 10-1074 0 6 12 18 24 30 36 0 50 100 150 200 250 Weeks post-AAV inoculation Antibody µg/mL rh2434 rh2436 rh2438 rh2443 anti-10-1074 0 6 12 18 24 30 36 0.0 0.5 1.0 1.5 2.0 Weeks post-AAV inoculation Absorbance at 450 nm rh2434 rh2436 rh2438 rh2443 10E8 0 6 12 18 24 30 36 0 50 100 150 200 250 Weeks post-AAV inoculation Antibody µg/mL rh2434 rh2436 rh2438 rh2443 anti-10E8 0 6 12 18 24 30 36 0.0 0.5 1.0 1.5 2.0 Weeks post-AAV inoculation Absorbance at 450 nm rh2434 rh2436 rh2438 rh2443 0 4 8 12 16 20 24 28 32 36 0 10 20 30 40 50 100 150 200 Weeks post-AAV Antibody µg/mL rh2438 antibody levels 3BNC117 10-1074 10E8 -100 -80 -60 -40 -20 20 40 60 80 SHIV-AD8 infection AAV administration 0.0 0.2 0.4 0.6 0.8 Weeks from AAV administration Absorbance at 450 nm Antibodies to gp120 0 -100 -80 -60 -40 -20 20 40 60 80 SHIV-AD8 infection AAV administration 0.0 0.5 1.0 1.5 Weeks from AAV administration Absorbance at 450 nm Antibodies to p27 0 0 10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 10 1 10 2 10 3 10 4 10 5 10 6 10 7 Weeks post-SHIV infection SHIV-AD8 RNA copies/mL in plasma SHIV-AD8 Viral loads rh2438 AAV administration 2. After AAV administration, animal rh2438 achieved profound and sustained virologic control during the chronic phase of the infection: 3. Monkey rh2438 persistently maintained high concentrations of broadly neutralizing anti-HIV antibodies 3BNC117 and 10-1074 in serum following the AAV administration: 1. Antibody and host anti-antibody levels were quantified by ELISA in all four AAV recipients: 5. Serum from rh2438 showed a sudden and marked decrease in antibody reactivity to p27/gag and g120/envelope after AAV inoculation : 4. Analysis of the viral reservoir in PBMCs isolated from rh2438 revealed a drop in both, viral RNA and viral DNA, after the AAV inoculation: 6. Attempts to recover SHIV from the peripheral blood of rh2438 did not yield any virus. Importantly, lymph node cells from this animal were not able to transmit infection when inoculated in two separate naïve recipient monkeys: HIV infection can be suppressed with anti-retroviral treatment (ART) but the existence of long-term reservoirs allows the viral load to rebound quickly after ART interruption 1 . Attempts to purge these reservoirs in vivo have not been successful and viral inducers may have serious undesirable effects 2,3 . Consequently, there is intense interest at the current time in finding strategies to prevent this rebound, i.e. finding a “functional cure”. Passive transfer of broadly neutralizing antibodies (bNAbs) can prevent infection 4-6 , and also suppress active infection in humanized mice and macaques 7-10 . Importantly, bNAbs can also suppress HIV emerging from the viral reservoir 11 and therefore they could be a good alternative to ART 12,13 . However, periodic administrations of large amounts of protein would be required for long-term effects, otherwise and similarly to ART, discontinuation of bNAb therapy would result in viral rebound 12 . A potential solution for overcoming this, is the use of recombinant adeno-associated virus vectors (AAV) 13 . AAV has an outstanding safety record in clinical trials 14 and, as long as the delivered protein is viewed as self 15 , it can result in continuous durable expression of the transgene product for years 16-20 . The idea is that HIV-infected people could get one shot of AAV making a cocktail of bNAbs and if satisfactory levels of antibody could be maintained in vivo, those individuals would remain suppressed for years without having to take ART or receive regular antibody administrations. Our intention is to perform experiments in monkeys that will inform and guide development of this concept for use in people. Methods Experimental infection with SHIV-AD8 (intravenous) Week 0 SHIV-AD8 infection AAV inoculation Week 86 Week 160 Week 164 1st Adoptive transfer 2nd Adoptive transfer Intramuscular inoculation of rAAV coding for anti-HIV antibodies 4 macaques After 86 weeks of SHIV-AD8 infection, four rhesus macaques were inoculated intramuscularly with three different AAVs coding for three different broadly neutralizing anti-HIV antibodies in a therapeutic approach: 10E8 3BNC117 10-1074 Four rhesus monkeys were infected with SHIV-AD8 for 86 weeks before receiving adeno-associated virus (AAV) vectors expressing rhesusized IgG1 versions of anti-HIV monoclonal antibodies 10E8, 3BNC117, and 10-1074 in a therapeutic approach. 10E8 was successfully delivered to none of the four monkeys; 3BNC117 to one of the four monkeys; 10-1074 to three of the four monkeys. SHIV-infected monkey rh2438 maintained serum concentrations of 3BNC117 and 10-1074 of 75 150 µg/ml for 17 months after AAV administration. Monkey rh2438 never received antiviral drugs at any time. The viral load set point in rh2438 was 10,000 20,000 viral RNA copies per ml of plasma for the prolonged period leading up to the time of AAV administration. Following the initial decline in viremia after AAV administration, viral loads remained suppressed to below the limit of detection of 30 RNA copies per ml of plasma for 17 months. During this time rh2438 anti-p27gag antibodies declined more than 10-fold. Attempts to recover SHIV from the peripheral blood of rh2438 were unsuccessful, including in vitro cultures of 10 8 CD8-depleted PBMCs. Two attempts to adoptively transfer infection to naïve recipients using lymph nodes taken from rh2438 failed to transmit the infection. These findings highlight the potential of AAV-mediated antibody expression for impacting HIV-1 infections. 0 10 20 30 40 50 60 70 80 1 10 100 1,000 Weeks from AAV administration RNA copies per 10 6 cells RNA 650 790 3.9 20 7.3 5.2 3 23 0 10 20 30 40 50 60 70 80 10 100 Weeks from AAV administration DNA copies per 10 6 cells DNA 88 57 22 20 26 25 16 23 Conclusions Viral loads were measured by RT-PCR. Antibody and anti-antibody levels were measured by standard ELISA. Adoptive transfer involved surgical removal of lymph nodes from the monkey with undetectable viremia, single cell suspension preparation and inoculation of the cells in two separate naïve recipient animals. 1. Chun, T.W. and A.S. Fauci, HIV reservoirs: pathogenesis and obstacles to viral eradication and cure. AIDS, 2012. 26(10): p. 1261-8. 2. Archin, N.M., et al., Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol, 2014. 12(11): p. 750-64. 3. Shan, L., et al., Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity, 2012. 36(3): p. 491-501. 4. Shingai, M., et al., Passive transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block SHIV infection in macaques. J Exp Med, 2014. 211(10): p. 2061-74. 5. Saunders, K.O., et al., Sustained Delivery of a Broadly Neutralizing Antibody in Nonhuman Primates Confers Long-Term Protection against Simian/Human Immunodeficiency Virus Infection. J Virol, 2015. 89(11): p. 5895-903. 6. Moldt, B., et al., Highly potent HIV-specific antibody neutralization in vitro translates into effective protection against mucosal SHIV challenge in vivo. Proc Natl Acad Sci U S A, 2012. 109(46): p. 18921-5. 7. Klein, F., et al., HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature, 2012. 492(7427): p. 118-22. 8. Barouch, D.H., et al., Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature, 2013. 503(7475): p. 224-8. 9. Shingai, M., et al., Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature, 2013. 503(7475): p. 277-80. 10. Horwitz, J.A., et al., HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice. Proc Natl Acad Sci U S A, 2013. 110(41): p. 16538-43. 11. Chun, T.W., et al., Broadly neutralizing antibodies suppress HIV in the persistent viral reservoir. Proc Natl Acad Sci U S A, 2014. 111(36): p. 13151-6. 12. Halper-Stromberg, A., et al., Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice. Cell, 2014. 158(5): p. 989-99. 13. Deal, C.E. and A.B. Balazs, Vectored antibody gene delivery for the prevention or treatment of HIV infection. Curr Opin HIV AIDS, 2015. 10(3): p. 190-7. 14. Hastie, E. and R.J. Samulski, Adeno-associated virus at 50: a golden anniversary of discovery, research, and gene therapy success-a personal perspective. Hum Gene Ther, 2015. 26(5): p. 257-65. 15. Martinez-Navio, J.M., et al., Host Anti-antibody Responses Following AAV-Mediated Delivery of Antibodies against HIV and SIV in Rhesus Monkeys. Mol Ther, 2015. 16. Daya, S. and K.I. Berns, Gene therapy using adeno-associated virus vectors. Clin Microbiol Rev, 2008. 21(4): p. 583-93. 17. Goncalves, M.A., Adeno-associated virus: from defective virus to effective vector. Virol J, 2005. 2: p. 43. 18. McCarty, D.M., Self-complementary AAV vectors; advances and applications. Mol Ther, 2008. 16(10): p. 1648-56. 19. Schultz, B.R. and J.S. Chamberlain, Recombinant adeno-associated virus transduction and integration. Mol Ther, 2008. 16(7): p. 1189-99. 20. Mingozzi, F. and K.A. High, Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat Rev Genet, 2011. 12(5): p. 341-55. 1. We have shown evidence for an actual cure of a SHIV-infected monkey during the chronic phase of the infection. 2. Long term delivery of potent and broadly neutralizing antibodies with AAV is a very promising approach against HIV. Surgical removal of lymph nodes and generation of single cell suspensions w160 rh2438 r11027 r07034 No virus !! w164 * *

Evidence for an Actual Cure of a SHIV-infected Monkeycfar.med.miami.edu/documents/Jose_Martinez-Navio.pdf · Evidence for an Actual Cure of a SHIV-infected Monkey Abstract Methods

Embed Size (px)

Citation preview

Evidence for an Actual Cure of a SHIV-infected Monkey

Abstract Methods Background

Results

José M. Martinez-Navio1*, Sebastian P. Fuchs1,2*, Shara N. Pantry1, Natasha N. Duggan3, William A. Lauer1, Eva G. Rakasz4, Brandon F. Keele5, Michel C. Nussenzweig6,7, Jeffrey D. Lifson5, Guangping Gao8 & Ronald C. Desrosiers1°

1 Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. 2 Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054 Germany. 3 Department of Molecular Cell and Developmental Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. 4 Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 53711, USA. 5 AIDS and Cancer Virus Program,

Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, USA. 6 Laboratory of Molecular Immunology and 7 Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA. 8 Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, 01655, USA.

*These authors contributed equally to this work. °Mentor

References

3BNC117

0 6 12 18 24 30 360

50

100

150

200

250

Weeks post-AAV inoculation

Ant

ibod

y µg

/mL

rh2434rh2436rh2438rh2443

anti-3BNC117

0 6 12 18 24 30 360.0

0.5

1.0

1.5

2.0

Weeks post-AAV inoculation

Abs

orba

nce

at 4

50 n

m

rh2434rh2436rh2438rh2443

10-1074

0 6 12 18 24 30 360

50

100

150

200

250

Weeks post-AAV inoculation

Ant

ibod

y µg

/mL

rh2434rh2436rh2438rh2443

anti-10-1074

0 6 12 18 24 30 360.0

0.5

1.0

1.5

2.0

Weeks post-AAV inoculation

Abs

orba

nce

at 4

50 n

m

rh2434rh2436rh2438rh2443

10E8

0 6 12 18 24 30 360

50

100

150

200

250

Weeks post-AAV inoculation

Ant

ibod

y µg

/mL

rh2434rh2436rh2438rh2443

anti-10E8

0 6 12 18 24 30 360.0

0.5

1.0

1.5

2.0

Weeks post-AAV inoculation

Abs

orba

nce

at 4

50 n

m

rh2434rh2436rh2438rh2443

0 4 8 12 16 20 24 28 32 360

10

20

30

40

50100150200

Weeks post-AAV

Ant

ibod

y µg

/mL

rh2438 antibody levels

3BNC11710-1074

10E8

-100 -80 -60 -40 -20 20 40 60 80

SHIV-AD8 infection AAV administration

0.0

0.2

0.4

0.6

0.8

Weeks from AAV administration

Abs

orba

nce

at 4

50 n

m

Antibodies to gp120

0-100 -80 -60 -40 -20 20 40 60 80

SHIV-AD8 infection AAV administration

0.0

0.5

1.0

1.5

Weeks from AAV administration

Abs

orba

nce

at 4

50 n

m

Antibodies to p27

0

0 10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160101

102

103

104

105

106

107

Weeks post-SHIV infection

SH

IV-A

D8

RN

A c

opie

s/m

L in

pla

sma

SHIV-AD8 Viral loadsrh2438

AAV administration

2. After AAV administration, animal rh2438 achieved profound and sustained virologic control during the chronic phase of the infection:

3. Monkey rh2438 persistently maintained high concentrations of broadly neutralizing anti-HIV antibodies 3BNC117 and 10-1074 in serum following the AAV administration:

1. Antibody and host anti-antibody levels were quantified by ELISA in all four AAV recipients:

5. Serum from rh2438 showed a sudden and marked decrease in antibody reactivity to p27/gag and g120/envelope after AAV inoculation :

4. Analysis of the viral reservoir in PBMCs isolated from rh2438 revealed a drop in both, viral RNA and viral DNA, after the AAV inoculation:

6. Attempts to recover SHIV from the peripheral blood of rh2438 did not yield any virus. Importantly, lymph node cells from this animal were not able to transmit infection when inoculated in two separate naïve recipient monkeys:

HIV infection can be suppressed with anti-retroviral treatment (ART) but the existence of long-term reservoirs allows the viral load to rebound quickly after ART interruption1. Attempts to purge these reservoirs in vivo have not been successful and viral inducers may have serious undesirable effects2,3. Consequently, there is intense interest at the current time in finding strategies to prevent this rebound, i.e. finding a “functional cure”. Passive transfer of broadly neutralizing antibodies (bNAbs) can prevent infection4-6, and also suppress active infection in humanized mice and macaques7-10. Importantly, bNAbs can also suppress HIV emerging from the viral reservoir11 and therefore they could be a good alternative to ART12,13. However, periodic administrations of large amounts of protein would be required for long-term effects, otherwise and similarly to ART, discontinuation of bNAb therapy would result in viral rebound12. A potential solution for overcoming this, is the use of recombinant adeno-associated virus vectors (AAV)13. AAV has an outstanding safety record in clinical trials14 and, as long as the delivered protein is viewed as self15, it can result in continuous durable expression of the transgene product for years16-20. The idea is that HIV-infected people could get one shot of AAV making a cocktail of bNAbs and if satisfactory levels of antibody could be maintained in vivo, those individuals would remain suppressed for years without having to take ART or receive regular antibody administrations. Our intention is to perform experiments in monkeys that will inform and guide development of this concept for use in people.

Methods

Experimental infection with SHIV-AD8 (intravenous)

Week 0

SHIV-AD8 infection

AAV inoculation

Week 86

Week 160

Week 164

1st Adoptive transfer

2nd Adoptive transfer

Intramuscular inoculation of rAAV coding for anti-HIV antibodies

4macaques

After 86 weeks of SHIV-AD8 infection, four rhesus macaques were inoculated intramuscularly with three different AAVs coding for three different broadly neutralizing anti-HIV antibodies in a therapeutic approach:

10E83BNC11710-1074

Four rhesus monkeys were infected with SHIV-AD8 for 86 weeks before receiving adeno-associated virus (AAV) vectors expressing rhesusized IgG1 versions of anti-HIV monoclonal antibodies 10E8, 3BNC117, and 10-1074 in a therapeutic approach. 10E8 was successfully delivered to none of the four monkeys; 3BNC117 to one of the four monkeys; 10-1074 to three of the four monkeys. SHIV-infected monkey rh2438 maintained serum concentrations of 3BNC117 and 10-1074 of 75 – 150 µg/ml for 17 months after AAV administration. Monkey rh2438 never received antiviral drugs at any time. The viral load set point in rh2438 was 10,000 – 20,000 viral RNA copies per ml of plasma for the prolonged period leading up to the time of AAV administration. Following the initial decline in viremia after AAV administration, viral loads remained suppressed to below the limit of detection of 30 RNA copies per ml of plasma for 17 months. During this time rh2438 anti-p27gag antibodies declined more than 10-fold. Attempts to recover SHIV from the peripheral blood of rh2438 were unsuccessful, including in vitro cultures of 108 CD8-depleted PBMCs. Two attempts to adoptively transfer infection to naïve recipients using lymph nodes taken from rh2438 failed to transmit the infection. These findings highlight the potential of AAV-mediated antibody expression for impacting HIV-1 infections.

0 10 20 30 40 50 60 70 801

10

100

1,000

Weeks from AAV administration

RN

A c

opie

s pe

r 106

cells

RNA

650

790

3.9

20

7.3

5.2

3

23

0 10 20 30 40 50 60 70 8010

100

Weeks from AAV administration

DN

A c

opie

s pe

r 106

cells

DNA88

57

2220

26 25

16

23

Conclusions

Viral loads were measured by RT-PCR. Antibody and anti-antibody levels were measured by standard ELISA. Adoptive transfer involved surgical removal of lymph nodes from the monkey with undetectable viremia, single cell suspension preparation and inoculation of the cells in two separate naïve recipient animals.

1. Chun, T.W. and A.S. Fauci, HIV reservoirs: pathogenesis and obstacles to viral eradication and cure. AIDS, 2012. 26(10): p. 1261-8. 2. Archin, N.M., et al., Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol, 2014. 12(11): p. 750-64. 3. Shan, L., et al., Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity, 2012. 36(3): p. 491-501. 4. Shingai, M., et al., Passive transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block SHIV infection in macaques. J Exp Med, 2014. 211(10): p. 2061-74. 5. Saunders, K.O., et al., Sustained Delivery of a Broadly Neutralizing Antibody in Nonhuman Primates Confers Long-Term Protection against Simian/Human Immunodeficiency Virus Infection. J Virol, 2015. 89(11): p. 5895-903. 6. Moldt, B., et al., Highly potent HIV-specific antibody neutralization in vitro translates into effective protection against mucosal SHIV challenge in vivo. Proc Natl Acad Sci U S A, 2012. 109(46): p. 18921-5. 7. Klein, F., et al., HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature, 2012. 492(7427): p. 118-22. 8. Barouch, D.H., et al., Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature, 2013. 503(7475): p. 224-8. 9. Shingai, M., et al., Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature, 2013. 503(7475): p. 277-80. 10. Horwitz, J.A., et al., HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice. Proc Natl Acad Sci U S A, 2013. 110(41): p. 16538-43. 11. Chun, T.W., et al., Broadly neutralizing antibodies suppress HIV in the persistent viral reservoir. Proc Natl Acad Sci U S A, 2014. 111(36): p. 13151-6. 12. Halper-Stromberg, A., et al., Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice. Cell, 2014. 158(5): p. 989-99. 13.  Deal, C.E. and A.B. Balazs, Vectored antibody gene delivery for the prevention or treatment of HIV infection. Curr Opin HIV AIDS, 2015. 10(3): p. 190-7. 14.  Hastie, E. and R.J. Samulski, Adeno-associated virus at 50: a golden anniversary of discovery, research, and gene therapy success-a personal perspective. Hum Gene Ther, 2015. 26(5): p. 257-65. 15. Martinez-Navio, J.M., et al., Host Anti-antibody Responses Following AAV-Mediated Delivery of Antibodies against HIV and SIV in Rhesus Monkeys. Mol Ther, 2015. 16. Daya, S. and K.I. Berns, Gene therapy using adeno-associated virus vectors. Clin Microbiol Rev, 2008. 21(4): p. 583-93. 17. Goncalves, M.A., Adeno-associated virus: from defective virus to effective vector. Virol J, 2005. 2: p. 43. 18. McCarty, D.M., Self-complementary AAV vectors; advances and applications. Mol Ther, 2008. 16(10): p. 1648-56. 19.  Schultz, B.R. and J.S. Chamberlain, Recombinant adeno-associated virus transduction and integration. Mol Ther, 2008. 16(7): p. 1189-99. 20.  Mingozzi, F. and K.A. High, Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat Rev Genet, 2011. 12(5): p. 341-55.

1. We have shown evidence for an actual cure of a SHIV-infected monkey during the chronic phase of the infection. 2. Long term delivery of potent and broadly neutralizing antibodies with AAV is a very promising approach against HIV.

Surgical removal of lymph nodes and generation of single cell suspensions

w160

rh2438 r11027

r07034

No virus !! w164

*

*