1
|
Pons-Faudoa FP, Di Trani N, Capuani S, Campa-Carranza JN, Nehete B, Sharma S, Shelton KA, Bushman LR, Abdelmawla F, Williams M, Roon L, Nerguizian D, Chua CYX, Ittmann MM, Nichols JE, Kimata JT, Anderson PL, Nehete PN, Arduino RC, Grattoni A. Long-acting refillable nanofluidic implant confers protection against SHIV infection in nonhuman primates. Sci Transl Med 2023; 15:eadg2887. [PMID: 37379369 DOI: 10.1126/scitranslmed.adg2887] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
The impact of pre-exposure prophylaxis (PrEP) on slowing the global HIV epidemic hinges on effective drugs and delivery platforms. Oral drug regimens are the pillar of HIV PrEP, but variable adherence has spurred development of long-acting delivery systems with the aim of increasing PrEP access, uptake, and persistence. We have developed a long-acting subcutaneous nanofluidic implant that can be refilled transcutaneously for sustained release of the HIV drug islatravir, a nucleoside reverse transcriptase translocation inhibitor that is used for HIV PrEP. In rhesus macaques, the islatravir-eluting implants achieved constant concentrations of islatravir in plasma (median 3.14 nM) and islatravir triphosphate in peripheral blood mononuclear cells (median 0.16 picomole per 106 cells) for more than 20 months. These drug concentrations were above the established PrEP protection threshold. In two unblinded, placebo-controlled studies, islatravir-eluting implants conferred 100% protection against infection with SHIVSF162P3 after repeated low-dose rectal or vaginal challenge in male or female rhesus macaques, respectively, compared to placebo control groups. The islatravir-eluting implants were well tolerated with mild local tissue inflammation and no signs of systemic toxicity over the 20-month study period. This refillable islatravir-eluting implant has potential as a long-acting drug delivery system for HIV PrEP.
Collapse
Affiliation(s)
- Fernanda P Pons-Faudoa
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Nicola Di Trani
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Simone Capuani
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- University of Chinese Academy of Science (UCAS), 19 Yuquan Road, Beijing 100049, China
| | - Jocelyn Nikita Campa-Carranza
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- School of Medicine and Health Sciences, Tecnológico de Monterrey, Monterrey, Mexico
| | - Bharti Nehete
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Suman Sharma
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kathryn A Shelton
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Lane R Bushman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Farah Abdelmawla
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Martin Williams
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laura Roon
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Nerguizian
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Corrine Ying Xuan Chua
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joan E Nichols
- Department of Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Pramod N Nehete
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA
- University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA
| | - Roberto C Arduino
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at University of Texas Health Science Center, Houston, TX 77030, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
2
|
Thippeshappa R, Polacino P, Chandrasekar SS, Truong K, Misra A, Aulicino PC, Hu SL, Kaushal D, Kimata JT. In vivo Serial Passaging of Human-Simian Immunodeficiency Virus Clones Identifies Characteristics for Persistent Viral Replication. Front Microbiol 2021; 12:779460. [PMID: 34867922 PMCID: PMC8636705 DOI: 10.3389/fmicb.2021.779460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
We previously reported that a human immunodeficiency virus type 1 with a simian immunodeficiency virus vif substitution (HSIV-vifNL4-3) could replicate in pigtailed macaques (PTMs), demonstrating that Vif is a species-specific tropism factor of primate lentiviruses. However, infections did not result in high-peak viremia or setpoint plasma viral loads, as observed during simian immunodeficiency virus (SIV) infection of PTMs. Here, we characterized variants isolated from one of the original infected animals with CD4 depletion after nearly 4years of infection to identify determinants of increased replication fitness. In our studies, we found that the HSIV-vif clones did not express the HIV-1 Vpr protein due to interference from the vpx open reading frame (ORF) in singly spliced vpr mRNA. To examine whether these viral genes contribute to persistent viral replication, we generated infectious HSIV-vif clones expressing either the HIV-1 Vpr or SIV Vpx protein. And then to determine viral fitness determinants of HSIV-vif, we conducted three rounds of serial in vivo passaging in PTMs, starting with an initial inoculum containing a mixture of CXCR4-tropic [Vpr-HSIV-vifNL4-3 isolated at 196 (C/196) and 200 (C/200) weeks post-infection from a PTM with depressed CD4 counts] and CCR5-tropic HSIV (Vpr+ HSIV-vif derivatives based NL-AD8 and Bru-Yu2 and a Vpx expressing HSIV-vifYu2). Interestingly, all infected PTMs showed peak plasma viremia close to or above 105 copies/ml and persistent viral replication for more than 20weeks. Infectious molecular clones (IMCs) recovered from the passage 3 PTM (HSIV-P3 IMCs) included mutations required for HIV-1 Vpr expression and those mutations encoded by the CXCR4-tropic HSIV-vifNL4-3 isolate C/196. The data indicate that the viruses selected during long-term infection acquired HIV-1 Vpr expression, suggesting the importance of Vpr for in vivo pathogenesis. Further passaging of HSIV-P3 IMCs in vivo may generate pathogenic variants with higher replication capacity, which will be a valuable resource as challenge virus in vaccine and cure studies.
Collapse
Affiliation(s)
- Rajesh Thippeshappa
- Disease Intervention and Prevention Program, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Patricia Polacino
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Shaswath S Chandrasekar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Khanghy Truong
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Anisha Misra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Paula C Aulicino
- Laboratorio de Biología Celular y Retrovirus, Hospital de Pediatría "Juan P. Garrahan"-CONICET, Buenos Aires, Argentina
| | - Shiu-Lok Hu
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States.,Department of Pharmaceutics, University of Washington, Seattle, WA, United States
| | - Deepak Kaushal
- Host-Pathogen Interactions Program, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
3
|
Pons-Faudoa FP, Sizovs A, Shelton KA, Momin Z, Niles JA, Bushman LR, Xu J, Chua CYX, Nichols JE, Demaria S, Ittmann MM, Hawkins T, Rooney JF, Marzinke MA, Kimata JT, Anderson PL, Nehete PN, Arduino RC, Ferrari M, Sastry KJ, Grattoni A. Preventive efficacy of a tenofovir alafenamide fumarate nanofluidic implant in SHIV-challenged nonhuman primates. ADVANCED THERAPEUTICS 2021; 4:2000163. [PMID: 33997267 PMCID: PMC8114879 DOI: 10.1002/adtp.202000163] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Indexed: 12/14/2022]
Abstract
Pre-exposure prophylaxis (PrEP) using antiretroviral oral drugs is effective at preventing HIV transmission when individuals adhere to the dosing regimen. Tenofovir alafenamide (TAF) is a potent antiretroviral drug, with numerous long-acting (LA) delivery systems under development to improve PrEP adherence. However, none has undergone preventive efficacy assessment. Here we show that LA TAF using a novel subcutaneous nanofluidic implant (nTAF) confers partial protection from HIV transmission. We demonstrate that sustained subcutaneous delivery through nTAF in rhesus macaques maintained tenofovir diphosphate concentration at a median of 390.00 fmol/106 peripheral blood mononuclear cells, 9 times above clinically protective levels. In a non-blinded, placebo-controlled rhesus macaque study with repeated low-dose rectal SHIVSF162P3 challenge, the nTAF cohort had a 62.50% reduction (95% CI: 1.72% to 85.69%; p=0.068) in risk of infection per exposure compared to the control. Our finding mirrors that of tenofovir disoproxil fumarate (TDF) monotherapy, where 60.00% protective efficacy was observed in macaques, and clinically, 67.00% reduction in risk with 86.00% preventive efficacy in individuals with detectable drug in the plasma. Overall, our nanofluidic technology shows potential as a subcutaneous delivery platform for long-term PrEP and provides insights for clinical implementation of LA TAF for HIV prevention.
Collapse
Affiliation(s)
- Fernanda P Pons-Faudoa
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Antons Sizovs
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Kathryn A Shelton
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Zoha Momin
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jean A Niles
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA
| | - Lane R Bushman
- Deparment of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado- Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jiaqiong Xu
- Center for Outcomes Research and DeBakey Heart and Vascular Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Corrine Ying Xuan Chua
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Joan E Nichols
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Mark A Marzinke
- Departments of Pathology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter L Anderson
- Deparment of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado- Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Pramod N Nehete
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Roberto C Arduino
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mauro Ferrari
- School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - K Jagannadha Sastry
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
4
|
Pons-Faudoa FP, Trani ND, Sizovs A, Shelton KA, Momin Z, Bushman LR, Xu J, Lewis DE, Demaria S, Hawkins T, Rooney JF, Marzinke MA, Kimata JT, Anderson PL, Nehete PN, Arduino RC, Sastry KJ, Grattoni A. Viral load Reduction in SHIV-Positive Nonhuman Primates via Long-Acting Subcutaneous Tenofovir Alafenamide Fumarate Release from a Nanofluidic Implant. Pharmaceutics 2020; 12:E981. [PMID: 33080776 PMCID: PMC7590004 DOI: 10.3390/pharmaceutics12100981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
HIV-1 is a chronic disease managed by strictly adhering to daily antiretroviral therapy (ART). However, not all people living with HIV-1 have access to ART, and those with access may not adhere to treatment regimens increasing viral load and disease progression. Here, a subcutaneous nanofluidic implant was used as a long-acting (LA) drug delivery platform to address these issues. The device was loaded with tenofovir alafenamide (TAF) and implanted in treatment-naïve simian HIV (SHIV)-positive nonhuman primates (NHP) for a month. We monitored intracellular tenofovir-diphosphate (TFV-DP) concentration in the target cells, peripheral blood mononuclear cells (PBMC). The concentrations of TFV-DP were maintained at a median of 391.0 fmol/106 cells (IQR, 243.0 to 509.0 fmol/106 cells) for the duration of the study. Further, we achieved drug penetration into lymphatic tissues, known for persistent HIV-1 replication. Moreover, we observed a first-phase viral load decay of -1.14 ± 0.81 log10 copies/mL (95% CI, -0.30 to -2.23 log10 copies/mL), similar to -1.08 log10 copies/mL decay observed in humans. Thus, LA TAF delivered from our nanofluidic implant had similar effects as oral TAF dosing with a lower dose, with potential as a platform for LA ART.
Collapse
Affiliation(s)
- Fernanda P. Pons-Faudoa
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (F.P.P.-F.); (N.D.T.); (A.S.)
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey 64710, NL, Mexico
| | - Nicola Di Trani
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (F.P.P.-F.); (N.D.T.); (A.S.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Science (UCAS), Shijingshan, Beijing 100049, China
| | - Antons Sizovs
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (F.P.P.-F.); (N.D.T.); (A.S.)
| | - Kathryn A. Shelton
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA; (K.A.S.); (P.N.N.); (K.J.S.)
| | - Zoha Momin
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (Z.M.); (J.T.K.)
| | - Lane R. Bushman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; (L.R.B.); (P.L.A.)
| | - Jiaqiong Xu
- Center for Outcomes Research and DeBakey Heart and Vascular Center, Houston Methodist Research Institute, Houston, TX 77030, USA;
- Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA;
- Department of Pathology and Laboratory of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Trevor Hawkins
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (T.H.); (J.F.R.)
| | - James F. Rooney
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (T.H.); (J.F.R.)
| | - Mark A. Marzinke
- Departments of Pathology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA;
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (Z.M.); (J.T.K.)
| | - Peter L. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; (L.R.B.); (P.L.A.)
| | - Pramod N. Nehete
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA; (K.A.S.); (P.N.N.); (K.J.S.)
- The University of Texas MD Anderson Cancer Center UTH Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Roberto C. Arduino
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - K. Jagannadha Sastry
- Department of Comparative Medicine, Michael E. Keeling Center for Comparative Medicine and Research, MD Anderson Cancer Center, Bastrop, TX 78602, USA; (K.A.S.); (P.N.N.); (K.J.S.)
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (F.P.P.-F.); (N.D.T.); (A.S.)
- Department of Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
5
|
Rust BJ, Kean LS, Colonna L, Brandenstein KE, Poole NH, Obenza W, Enstrom MR, Maldini CR, Ellis GI, Fennessey CM, Huang ML, Keele BF, Jerome KR, Riley JL, Kiem HP, Peterson CW. Robust expansion of HIV CAR T cells following antigen boosting in ART-suppressed nonhuman primates. Blood 2020; 136:1722-1734. [PMID: 32614969 PMCID: PMC7544543 DOI: 10.1182/blood.2020006372] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19+ hematologic malignancies have rapidly emerged as a promising, novel therapy. In contrast, results from the few CAR T-cell studies for infectious diseases such as HIV-1 have been less convincing. These challenges are likely due to the low level of antigen present in antiretroviral therapy (ART)-suppressed patients in contrast to those with hematologic malignancies. Using our well-established nonhuman primate model of ART-suppressed HIV-1 infection, we tested strategies to overcome these limitations and challenges. We first optimized CAR T-cell production to maintain central memory subsets, consistent with current clinical paradigms. We hypothesized that additional exogenous antigen might be required in an ART-suppressed setting to aid expansion and persistence of CAR T cells. Thus, we studied 4 simian/HIV-infected, ART-suppressed rhesus macaques infused with virus-specific CD4CAR T cells, followed by supplemental infusion of cell-associated HIV-1 envelope (Env). Env boosting led to significant and unprecedented expansion of virus-specific CAR+ T cells in vivo; after ART treatment interruption, viral rebound was significantly delayed compared with controls (P = .014). In 2 animals with declining CAR T cells, rhesusized anti-programmed cell death protein 1 (PD-1) antibody was administered to reverse PD-1-dependent immune exhaustion. Immune checkpoint blockade triggered expansion of exhausted CAR T cells and concordantly lowered viral loads to undetectable levels. These results show that supplemental cell-associated antigen enables robust expansion of CAR T cells in an antigen-sparse environment. To our knowledge, this is the first study to show expansion of virus-specific CAR T cells in infected, suppressed hosts, and delay/control of viral recrudescence.
Collapse
Affiliation(s)
- Blake J Rust
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Leslie S Kean
- Boston Children's Hospital/Dana-Farber Cancer Institute-Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Lucrezia Colonna
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Nikhita H Poole
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Willimark Obenza
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mark R Enstrom
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Colby R Maldini
- Department of Microbiology and Center for Cellular Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gavin I Ellis
- Department of Microbiology and Center for Cellular Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Meei-Li Huang
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Keith R Jerome
- Department of Laboratory Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
| | - James L Riley
- Department of Microbiology and Center for Cellular Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine, University of Washington, Seattle, WA
| | - Christopher W Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
6
|
Evidence for persistence of the SHIV reservoir early after MHC haploidentical hematopoietic stem cell transplantation. Nat Commun 2018; 9:4438. [PMID: 30361514 PMCID: PMC6202377 DOI: 10.1038/s41467-018-06736-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/19/2018] [Indexed: 01/08/2023] Open
Abstract
Allogeneic transplantation (allo-HCT) has led to the cure of HIV in one individual, raising the question of whether transplantation can eradicate the HIV reservoir. To test this, we here present a model of allo-HCT in SHIV-infected, cART-suppressed nonhuman primates. We infect rhesus macaques with SHIV-1157ipd3N4, suppress them with cART, then transplant them using MHC-haploidentical allogeneic donors during continuous cART. Transplant results in ~100% myeloid donor chimerism, and up to 100% T-cell chimerism. Between 9 and 47 days post-transplant, terminal analysis shows that while cell-associated SHIV DNA levels are reduced in the blood and in lymphoid organs post-transplant, the SHIV reservoir persists in multiple organs, including the brain. Sorting of donor-vs.-recipient cells reveals that this reservoir resides in recipient cells. Moreover, tetramer analysis indicates a lack of virus-specific donor immunity post-transplant during continuous cART. These results suggest that early post-transplant, allo-HCT is insufficient for recipient reservoir eradication despite high-level donor chimerism and GVHD. Allogeneic hematopoietic cell transplantation (allo-HCT) has led to the cure of HIV in one individual, but the underlying mechanisms are unclear. Here, the authors present a model of allo-HCT in SHIV-infected nonhuman primates and show that the SHIV reservoir persists in multiple tissues early after transplantation.
Collapse
|
7
|
Peterson CW, Wang J, Deleage C, Reddy S, Kaur J, Polacino P, Reik A, Huang ML, Jerome KR, Hu SL, Holmes MC, Estes JD, Kiem HP. Differential impact of transplantation on peripheral and tissue-associated viral reservoirs: Implications for HIV gene therapy. PLoS Pathog 2018; 14:e1006956. [PMID: 29672640 PMCID: PMC5908070 DOI: 10.1371/journal.ppat.1006956] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
Abstract
Autologous transplantation and engraftment of HIV-resistant cells in sufficient numbers should recapitulate the functional cure of the Berlin Patient, with applicability to a greater number of infected individuals and with a superior safety profile. A robust preclinical model of suppressed HIV infection is critical in order to test such gene therapy-based cure strategies, both alone and in combination with other cure strategies. Here, we present a nonhuman primate (NHP) model of latent infection using simian/human immunodeficiency virus (SHIV) and combination antiretroviral therapy (cART) in pigtail macaques. We demonstrate that transplantation of CCR5 gene-edited hematopoietic stem/progenitor cells (HSPCs) persist in infected and suppressed animals, and that protected cells expand through virus-dependent positive selection. CCR5 gene-edited cells are readily detectable in tissues, namely those closely associated with viral reservoirs such as lymph nodes and gastrointestinal tract. Following autologous transplantation, tissue-associated SHIV DNA and RNA levels in suppressed animals are significantly reduced (p ≤ 0.05), relative to suppressed, untransplanted control animals. In contrast, the size of the peripheral reservoir, measured by QVOA, is variably impacted by transplantation. Our studies demonstrate that CCR5 gene editing is equally feasible in infected and uninfected animals, that edited cells persist, traffic to, and engraft in tissue reservoirs, and that this approach significantly reduces secondary lymphoid tissue viral reservoir size. Our robust NHP model of HIV gene therapy and viral persistence can be immediately applied to the investigation of combinatorial approaches that incorporate anti-HIV gene therapy, immune modulators, therapeutic vaccination, and latency reversing agents.
Collapse
Affiliation(s)
- Christopher W. Peterson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Jianbin Wang
- Sangamo Therapeutics, Richmond, CA, United States of America
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States of America
| | - Sowmya Reddy
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jasbir Kaur
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Patricia Polacino
- Washington National Primate Research Center, Seattle, WA, United States of America
| | - Andreas Reik
- Sangamo Therapeutics, Richmond, CA, United States of America
| | - Meei-Li Huang
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Keith R. Jerome
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America
| | - Shiu-Lok Hu
- Washington National Primate Research Center, Seattle, WA, United States of America
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
| | | | - Jacob D. Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States of America
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- Department of Pathology, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
8
|
Interferon-Inducible CD169/Siglec1 Attenuates Anti-HIV-1 Effects of Alpha Interferon. J Virol 2017; 91:JVI.00972-17. [PMID: 28794041 DOI: 10.1128/jvi.00972-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023] Open
Abstract
A hallmark of human immunodeficiency virus type 1 (HIV-1) infection in vivo is chronic immune activation concomitant with type I interferon (IFN) production. Although type I IFN induces an antiviral state in many cell types, HIV-1 can replicate in vivo via mechanisms that have remained unclear. We have recently identified a type I IFN-inducible protein, CD169, as the HIV-1 attachment factor on dendritic cells (DCs) that can mediate robust infection of CD4+ T cells in trans Since CD169 expression on macrophages is also induced by type I IFN, we hypothesized that type I IFN-inducible CD169 could facilitate productive HIV-1 infection in myeloid cells in cis and CD4+ T cells in trans and thus offset antiviral effects of type I IFN. In support of this hypothesis, infection of HIV-1 or murine leukemia virus Env (MLV-Env)-pseudotyped HIV-1 particles was enhanced in IFN-α-treated THP-1 monocytoid cells, and this enhancement was primarily dependent on CD169-mediated enhancement at the virus entry step, a phenomenon phenocopied in HIV-1 infections of IFN-α-treated primary monocyte-derived macrophages (MDMs). Furthermore, expression of CD169, a marker of type I IFN-induced immune activation in vivo, was enhanced in lymph nodes from pigtailed macaques infected with simian immunodeficiency virus (SIV) carrying HIV-1 reverse transcriptase (RT-SHIV), compared to uninfected macaques, and interestingly, there was extensive colocalization of p27gag and CD169, suggesting productive infection of CD169+ myeloid cells in vivo While cell-free HIV-1 infection of IFN-α-treated CD4+ T cells was robustly decreased, initiation of infection in trans via coculture with CD169+ IFN-α-treated DCs restored infection, suggesting that HIV-1 exploits CD169 in cis and in trans to attenuate a type I IFN-induced antiviral state.IMPORTANCE HIV-1 infection in humans causes immune activation characterized by elevated levels of proinflammatory cytokines, including type I interferons (IFN). Although type I IFN induces an antiviral state in many cell types in vitro, HIV-1 can replicate in vivo via mechanisms that have remained unclear. In this study, we tested the hypothesis that CD169, a type I IFN-inducible HIV-1 attachment factor, offsets antiviral effects of type I IFN. Infection of HIV-1 was rescued in IFN-α-treated myeloid cells via upregulation of CD169 and a subsequent increase in CD169-dependent virus entry. Furthermore, extensive colocalization of viral Gag and CD169 was observed in lymph nodes of infected pigtailed macaques, suggesting productive infection of CD169+ cells in vivo Treatment of dendritic cell (DC)-T cell cocultures with IFN-α upregulated CD169 expression on DCs and rescued HIV-1 infection of CD4+ T cells in trans, suggesting that HIV-1 exploits CD169 to attenuate type I IFN-induced restrictions.
Collapse
|
9
|
Reeves DB, Peterson CW, Kiem HP, Schiffer JT. Autologous Stem Cell Transplantation Disrupts Adaptive Immune Responses during Rebound Simian/Human Immunodeficiency Virus Viremia. J Virol 2017; 91:e00095-17. [PMID: 28404854 PMCID: PMC5469274 DOI: 10.1128/jvi.00095-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Primary HIV-1 infection induces a virus-specific adaptive/cytolytic immune response that impacts the plasma viral load set point and the rate of progression to AIDS. Combination antiretroviral therapy (cART) suppresses plasma viremia to undetectable levels that rebound upon cART treatment interruption. Following cART withdrawal, the memory component of the virus-specific adaptive immune response may improve viral control compared to primary infection. Here, using primary infection and treatment interruption data from macaques infected with simian/human immunodeficiency virus (SHIV), we observe a lower peak viral load but an unchanged viral set point during viral rebound. The addition of an autologous stem cell transplant before cART withdrawal alters viral dynamics: we found a higher rebound set point but similar peak viral loads compared to the primary infection. Mathematical modeling of the data that accounts for fundamental immune parameters achieves excellent fit to heterogeneous viral loads. Analysis of model output suggests that the rapid memory immune response following treatment interruption does not ultimately lead to better viral containment. Transplantation decreases the durability of the adaptive immune response following cART withdrawal and viral rebound. Our model's results highlight the impact of the endogenous adaptive immune response during primary SHIV infection. Moreover, because we capture adaptive immune memory and the impact of transplantation, this model will provide insight into further studies of cure strategies inspired by the Berlin patient.IMPORTANCE HIV patients who interrupt combination antiretroviral therapy (cART) eventually experience viral rebound, the return of viral loads to pretreatment levels. However, the "Berlin patient" remained free of HIV rebound over a decade after stopping cART. His cure is attributed to leukemia treatment that included an HIV-resistant stem cell transplant. Inspired by this case, we studied the impact of stem cell transplantation in a macaque simian/HIV (SHIV) system. Using a mechanistic mathematical model, we found that while primary infection generates an adaptive immune memory response, stem cell transplantation disrupts this learned immunity. The results have implications for HIV cure regimens based on stem cell transplantation.
Collapse
Affiliation(s)
- Daniel B Reeves
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Christopher W Peterson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Feder AF, Kline C, Polacino P, Cottrell M, Kashuba ADM, Keele BF, Hu SL, Petrov DA, Pennings PS, Ambrose Z. A spatio-temporal assessment of simian/human immunodeficiency virus (SHIV) evolution reveals a highly dynamic process within the host. PLoS Pathog 2017; 13:e1006358. [PMID: 28542550 PMCID: PMC5444849 DOI: 10.1371/journal.ppat.1006358] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/17/2017] [Indexed: 12/25/2022] Open
Abstract
The process by which drug-resistant HIV-1 arises and spreads spatially within an infected individual is poorly understood. Studies have found variable results relating how HIV-1 in the blood differs from virus sampled in tissues, offering conflicting findings about whether HIV-1 throughout the body is homogeneously distributed. However, most of these studies sample only two compartments and few have data from multiple time points. To directly measure how drug resistance spreads within a host and to assess how spatial structure impacts its emergence, we examined serial sequences from four macaques infected with RT-SHIVmne027, a simian immunodeficiency virus encoding HIV-1 reverse transcriptase (RT), and treated with RT inhibitors. Both viral DNA and RNA (vDNA and vRNA) were isolated from the blood (including plasma and peripheral blood mononuclear cells), lymph nodes, gut, and vagina at a median of four time points and RT was characterized via single-genome sequencing. The resulting sequences reveal a dynamic system in which vRNA rapidly acquires drug resistance concomitantly across compartments through multiple independent mutations. Fast migration results in the same viral genotypes present across compartments, but not so fast as to equilibrate their frequencies immediately. The blood and lymph nodes were found to be compartmentalized rarely, while both the blood and lymph node were more frequently different from mucosal tissues. This study suggests that even oft-sampled blood does not fully capture the viral dynamics in other parts of the body, especially the gut where vRNA turnover was faster than the plasma and vDNA retained fewer wild-type viruses than other sampled compartments. Our findings of transient compartmentalization across multiple tissues may help explain the varied results of previous compartmentalization studies in HIV-1.
Collapse
Affiliation(s)
- Alison F. Feder
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Christopher Kline
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Patricia Polacino
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Mackenzie Cottrell
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Angela D. M. Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Shiu-Lok Hu
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Dmitri A. Petrov
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Pleuni S. Pennings
- Department of Biology, San Francisco State University, San Francisco, CA, United States
| | - Zandrea Ambrose
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Peterson CW, Benne C, Polacino P, Kaur J, McAllister CE, Filali-Mouhim A, Obenza W, Pecor TA, Huang ML, Baldessari A, Murnane RD, Woolfrey AE, Jerome KR, Hu SL, Klatt NR, DeRosa S, Sékaly RP, Kiem HP. Loss of immune homeostasis dictates SHIV rebound after stem-cell transplantation. JCI Insight 2017; 2:e91230. [PMID: 28239658 DOI: 10.1172/jci.insight.91230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The conditioning regimen used as part of the Berlin patient's hematopoietic cell transplant likely contributed to his eradication of HIV infection. We studied the impact of conditioning in simian-human immunodeficiency virus-infected (SHIV-infected) macaques suppressed by combination antiretroviral therapy (cART). The conditioning regimen resulted in a dramatic, but incomplete depletion of CD4+ and CD8+ T cells and CD20+ B cells, increased T cell activation and exhaustion, and a significant loss of SHIV-specific Abs. The disrupted T cell homeostasis and markers of microbial translocation positively correlated with an increased viral rebound after cART interruption. Quantitative viral outgrowth and Tat/rev-induced limiting dilution assays showed that the size of the latent SHIV reservoir did not correlate with viral rebound. These findings identify perturbations of the immune system as a mechanism for the failure of autologous transplantation to eradicate HIV. Thus, transplantation strategies may be improved by incorporating immune modulators to prevent disrupted homeostasis, and gene therapy to protect transplanted cells.
Collapse
Affiliation(s)
- Christopher W Peterson
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Clarisse Benne
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Patricia Polacino
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Jasbir Kaur
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Cristina E McAllister
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Willi Obenza
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tiffany A Pecor
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Meei-Li Huang
- Division of Vaccine and Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Audrey Baldessari
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Robert D Murnane
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Ann E Woolfrey
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Keith R Jerome
- Division of Vaccine and Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Laboratory Medicine
| | - Shiu-Lok Hu
- Washington National Primate Research Center, Seattle, Washington, USA.,Department of Pharmaceutics and
| | - Nichole R Klatt
- Washington National Primate Research Center, Seattle, Washington, USA.,Department of Pharmaceutics and
| | - Stephen DeRosa
- Division of Vaccine and Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Rafick P Sékaly
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hans-Peter Kiem
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Peterson CW, Haworth KG, Burke BP, Polacino P, Norman KK, Adair JE, Hu SL, Bartlett JS, Symonds GP, Kiem HP. Multilineage polyclonal engraftment of Cal-1 gene-modified cells and in vivo selection after SHIV infection in a nonhuman primate model of AIDS. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16007. [PMID: 26958575 PMCID: PMC4765711 DOI: 10.1038/mtm.2016.7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/11/2022]
Abstract
We have focused on gene therapy approaches to induce functional cure/remission of HIV-1 infection. Here, we evaluated the safety and efficacy of the clinical grade anti-HIV lentiviral vector, Cal-1, in pigtailed macaques (Macaca nemestrina). Cal-1 animals exhibit robust levels of gene marking in myeloid and lymphoid lineages without measurable adverse events, suggesting that Cal-1 transduction and autologous transplantation of hematopoietic stem cells are safe, and lead to long-term, multilineage engraftment following myeloablative conditioning. Ex vivo, CD4+ cells from transplanted animals undergo positive selection in the presence of simian/human immunodeficiency virus (SHIV). In vivo, Cal-1 gene-marked cells are evident in the peripheral blood and in HIV-relevant tissue sites such as the gastrointestinal tract. Positive selection for gene-marked cells is observed in blood and tissues following SHIV challenge, leading to maintenance of peripheral blood CD4+ T-cell counts in a normal range. Analysis of Cal-1 lentivirus integration sites confirms polyclonal engraftment of gene-marked cells. Following infection, a polyclonal, SHIV-resistant clonal repertoire is established. These findings offer strong preclinical evidence for safety and efficacy of Cal-1, present a new method for tracking protected cells over the course of virus-mediated selective pressure in vivo, and reveal previously unobserved dynamics of virus-dependent T-cell selection.
Collapse
Affiliation(s)
- Christopher W Peterson
- Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, Washington, USA
| | - Kevin G Haworth
- Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, Washington, USA
| | | | - Patricia Polacino
- Washington National Primate Research Center , Seattle, Washington, USA
| | - Krystin K Norman
- Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, Washington, USA
| | - Jennifer E Adair
- Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, Washington, USA
| | - Shiu-Lok Hu
- Washington National Primate Research Center, Seattle, Washington, USA; Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | | | | | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
13
|
Oral Immunization with Recombinant Vaccinia Virus Prime and Intramuscular Protein Boost Provides Protection against Intrarectal Simian-Human Immunodeficiency Virus Challenge in Macaques. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 23:204-12. [PMID: 26718849 DOI: 10.1128/cvi.00597-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/23/2015] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) acquisition occurs predominantly through mucosal transmission. We hypothesized that greater mucosal immune responses and protective efficacy against mucosal HIV-1 infection may be achieved by prime-boost immunization at mucosal sites. We used a macaque model to determine the safety, immunogenicity, and protective efficacy of orally delivered, replication-competent but attenuated recombinant vaccinia viruses expressing full-length HIV-1 SF162 envelope (Env) or simian immunodeficiency virus (SIV) Gag-Pol proteins. We examined the dose and route that are suitable for oral immunization with recombinant vaccinia viruses. We showed that sublingual inoculation of two vaccinia virus-naive pigtailed macaques with 5 × 10(8) PFU of recombinant vaccinia viruses was safe. However, sublingual inoculation with a higher dose or tonsillar inoculation resulted in secondary oral lesions, indicating the need to optimize the dose and route for oral immunization with replication-competent vaccinia virus vectors. Oral priming alone elicited antibody responses to vaccinia virus and to the SF162 Env protein. Intramuscular immunization with the SF162 gp120 protein at either 20 or 21 weeks postpriming resulted in a significant boost in antibody responses in both systemic and mucosal compartments. Furthermore, we showed that immune responses induced by recombinant vaccinia virus priming and intramuscular protein boosting provided protection against intrarectal challenge with the simian-human immunodeficiency virus SHIV-SF162-P4.
Collapse
|
14
|
Peterson CW, Haworth KG, Polacino P, Huang ML, Sykes C, Obenza WM, Repetto AC, Kashuba A, Bumgarner R, DeRosa SC, Woolfrey AE, Jerome KR, Mullins JI, Hu SL, Kiem HP. Lack of viral control and development of combination antiretroviral therapy escape mutations in macaques after bone marrow transplantation. AIDS 2015; 29:1597-606. [PMID: 26372270 PMCID: PMC4572605 DOI: 10.1097/qad.0000000000000702] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We have previously demonstrated robust control of simian/human immunodeficiency virus (SHIV1157-ipd3N4) viremia following administration of combination antiretroviral therapy (cART) in pigtailed macaques. Here, we sought to determine the safety of hematopoietic stem cell transplantation (HSCT) in cART-suppressed and unsuppressed animals. DESIGN We compared disease progression in animals challenged with SHIV 100 days post-transplant, to controls that underwent transplant following SHIV challenge and stable cART-dependent viral suppression. METHODS SHIV viral load, cART levels, and anti-SHIV antibodies were measured longitudinally from plasma/serum from each animal. Flow cytometry was used to assess T-cell subset frequencies in peripheral blood and the gastrointestinal tract. Deep sequencing was used to identify cART resistance mutations. RESULTS In control animals, virus challenge induced transient peak viremia, viral set point, and durable suppression by cART. Subsequent HSCT was not associated with adverse events in these animals. Post-transplant animals were challenged during acute recovery following HSCT, and displayed sustained peak viremia and cART resistance. Although post-transplant animals had comparable plasma levels of antiretroviral drugs and showed no evidence of enhanced infection of myeloid subsets in the periphery, they exhibited a drastic reduction in virus-specific antibody production and decreased T-cell counts. CONCLUSIONS These results suggest that virus challenge prior to complete transplant recovery impairs viral control and may promote drug resistance. These findings may also have implications for scheduled treatment interruption studies in patients on cART during post-HSCT recovery: premature scheduled treatment interruption could similarly result in lack of viral control and cART resistance.
Collapse
Affiliation(s)
- Christopher W Peterson
- aClinical Research Division, Fred Hutchinson Cancer Research Center bWashington National Primate Research Center, Seattle cVaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington dDivision of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, North Carolina eDepartment of Microbiology fDepartment of Pediatrics gDepartment of Laboratory Medicine hDepartment of Medicine iDepartment of Pharmaceutics jDepartment of Pathology, University of Washington, Seattle, Washington, USA. *Christopher W. Peterson and Kevin G. Haworth contributed equally to the writing of this article
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Lentivirus-mediated Gene Transfer in Hematopoietic Stem Cells Is Impaired in SHIV-infected, ART-treated Nonhuman Primates. Mol Ther 2015; 23:943-951. [PMID: 25648264 DOI: 10.1038/mt.2015.19] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022] Open
Abstract
Recent studies have demonstrated that genetically modified hematopoietic stem cells (HSCs) can reduce HIV viremia. We have developed an HIV/AIDS-patient model in Simian/human immunodeficiency virus (SHIV)-infected pigtailed macaques that are stably suppressed on antiretroviral therapy (ART: raltegravir, emtricitabine and tenofovir). Following SHIV infection and ART, animals undergo autologous HSC transplantation (HSCT) with lentivirally transduced cluster of differentiation (CD)34(+) cells expressing the mC46 anti-HIV fusion protein. We show that SHIV(+), ART-treated animals had very low gene marking levels after HSCT. Pretransduction CD34(+) cells contained detectable levels of all three ART drugs, likely contributing to the low gene transfer efficiency. Following HSCT recovery and the cessation of ART, plasma viremia rebounded, indicating that myeloablative total body irradiation cannot completely eliminate viral reservoirs after autologous HSCT. The kinetics of recovery following autologous HSCT in SHIV(+), ART-treated macaques paralleled those observed following transplantation of control animals. However, T-cell subset analyses demonstrated a high percentage of C-C chemokine receptor 5 (CCR5)-expressing CD4(+) T-cells after HSCT. These data suggest that an extended ART interruption time may be required for more efficient lentiviral transduction. To avoid complications associated with ART interruption in the context of high percentages of CD4(+)CCR5(+)T-cells after HSCT, the use of vector systems not impaired by the presence of residual ART may also be beneficial.
Collapse
|
16
|
Barrenas F, Palermo RE, Agricola B, Agy MB, Aicher L, Carter V, Flanary L, Green RR, McLain R, Li Q, Lu W, Murnane R, Peng X, Thomas MJ, Weiss JM, Anderson DM, Katze MG. Deep transcriptional sequencing of mucosal challenge compartment from rhesus macaques acutely infected with simian immunodeficiency virus implicates loss of cell adhesion preceding immune activation. J Virol 2014; 88:7962-72. [PMID: 24807713 PMCID: PMC4097788 DOI: 10.1128/jvi.00543-14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/25/2014] [Indexed: 12/27/2022] Open
Abstract
Pathology resulting from human immunodeficiency virus (HIV) infection is driven by protracted inflammation; the primary loss of CD4(+) T cells is caused by activation-driven apoptosis. Recent studies of nonhuman primates (NHPs) have suggested that during the acute phase of infection, antiviral mucosal immunity restricts viral replication in the primary infection compartment. These studies imply that HIV achieves systemic infection as a consequence of a failure in host antiviral immunity. Here, we used high-dose intrarectal inoculation of rhesus macaques with simian immunodeficiency virus (SIV) SIVmac251 to examine how the mucosal immune system is overcome by SIV during acute infection. The host response in rectal mucosa was characterized by deep mRNA sequencing (mRNA-seq) at 3 and 12 days postinoculation (dpi) in 4 animals for each time point. While we observed a strong host transcriptional response at 3 dpi, functions relating to antiviral immunity were absent. Instead, we observed a significant number of differentially expressed genes relating to cell adhesion and reorganization of the cytoskeleton. We also observed downregulation of genes encoding members of the claudin family of cell adhesion molecules, which are coexpressed with genes associated with pathology in the colorectal mucosa, and a large number of noncoding transcripts. In contrast, at 12 dpi the differentially expressed genes were enriched in those involved with immune system functions, in particular, functions relating to T cells, B cells, and NK cells. Our findings indicate that host responses that negatively affect mucosal integrity occur before inflammation. Consequently, when inflammation is activated at peak viremia, mucosal integrity is already compromised, potentially enabling rapid tissue damage, driving further inflammation. Importance: The HIV pandemic is one of the major threats to human health, causing over a million deaths per year. Recent studies have suggested that mucosal antiviral immune responses play an important role in preventing systemic infection after exposure to the virus. Yet, despite their potential role in decreasing transmission rates between individuals, these antiviral mechanisms are poorly understood. Here, we carried out the first deep mRNA sequencing analysis of mucosal host responses in the primary infection compartment during acute SIV infection. We found that during acute infection, a significant host response was mounted in the mucosa before inflammation was triggered. Our analysis indicated that the response has a detrimental effect on tissue integrity, causing increased permeability, tissue damage, and recruitment of SIV target cells. These results emphasize the importance of mucosal host responses preceding immune activation in preventing systemic SIV infection.
Collapse
Affiliation(s)
- Fredrik Barrenas
- Department of Microbiology, University of Washington, Seattle, Washington, USA Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Robert E Palermo
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Brian Agricola
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Michael B Agy
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Lauri Aicher
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Victoria Carter
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Leon Flanary
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Richard R Green
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Randy McLain
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Wuxun Lu
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Robert Murnane
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Xinxia Peng
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Matthew J Thomas
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Jeffrey M Weiss
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - David M Anderson
- Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA Washington National Primate Research Center, University of Washington, Seattle, Washington, USA
| |
Collapse
|
17
|
Ambrose Z, Kline C, Polacino P, Hu SL. Dysregulation of multiple inflammatory molecules in lymph node and ileum of macaques during RT-SHIV infection with or without antiretroviral therapy. J Med Primatol 2014; 43:298-309. [PMID: 24784552 DOI: 10.1111/jmp.12126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND Pathogenic infection with human immunodeficiency virus type 1 (HIV-1) or simian immunodeficiency virus (SIV) is characterized by a loss of CD4+ T cells and chronic lymphocyte activation even during suppressive antiretroviral therapy (ART). METHODS Using NanoString, expression of >100 molecules associated with inflammation or immune activation was evaluated in mesenteric lymph nodes and ileum of macaques infected with a pathogenic SIV/HIV virus, RT-SHIV, during viremia or during suppressive ART and compared to uninfected controls. RESULTS Of the 105 RNA species quantified in the tissues, expression of 33 genes was altered significantly in one or both tissues during viremia but returned to normal levels during ART. However, 29 additional genes were altered in expression levels in the tissues of both viremic and ART-suppressed macaques. CONCLUSIONS Identification of the mechanisms of chronic inflammation in specific cells and in different tissues may help determine whether early ART initiation or novel therapies can prevent it.
Collapse
Affiliation(s)
- Zandrea Ambrose
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
18
|
Peterson CW, Younan P, Polacino PS, Maurice NJ, Miller HW, Prlic M, Jerome KR, Woolfrey AE, Hu SL, Kiem HP. Robust suppression of env-SHIV viremia in Macaca nemestrina by 3-drug ART is independent of timing of initiation during chronic infection. J Med Primatol 2014; 42:237-46. [PMID: 24025078 DOI: 10.1111/jmp.12060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nonhuman primates (NHPs) are an important model organism for studies of HIV pathogenesis and preclinical evaluation of anti-HIV therapies. The successful translation of NHP-derived data to clinically relevant anti-HIV studies will require better understanding of the viral strains and NHP species used and their responses to existing antiretroviral therapies (ART). METHODS Five pigtailed macaques (Macaca nemestrina) were productively infected with the SIV/HIV chimeric virus SHIV-1157 ipd3N4 following intravenous challenge. After 8 or 27 weeks, ART (PMPA, FTC, raltegravir) was initiated. Viral load, T-cell counts, and production of SHIV-specific antibodies were monitored throughout the course of infection and ART. RESULTS ART led to a rapid and sustained decrease in plasma viral load. Suppression of plasma viremia by ART was independent of the timing of initiation during chronic infection. CONCLUSIONS We present a new NHP model of HIV infection on antiretroviral therapy, which should prove applicable to multiple clinically relevant anti-HIV approaches.
Collapse
|
19
|
Vif substitution enables persistent infection of pig-tailed macaques by human immunodeficiency virus type 1. J Virol 2011; 85:3767-79. [PMID: 21289128 DOI: 10.1128/jvi.02438-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Among Old World monkeys, pig-tailed macaques (Pt) are uniquely susceptible to human immunodeficiency virus type 1 (HIV-1), although the infection does not persist. We demonstrate that the susceptibility of Pt T cells to HIV-1 infection is due to the absence of postentry inhibition by a TRIM5 isoform. Notably, substitution of the viral infectivity factor protein, Vif, with that from pathogenic SIVmne enabled replication of HIV-1 in Pt T cells in vitro. When inoculated into juvenile pig-tailed macaques, the Pt-tropic HIV-1 persistently replicated for more than 1.5 to 2 years, producing low but measurable plasma viral loads and persistent proviral DNA in peripheral blood mononuclear cells. It also elicited strong antibody responses. However, there was no decline in CD4(+) T cells or evidence of disease. Surprisingly, the Pt-tropic HIV-1 was rapidly controlled when inoculated into newborn Pt macaques, although it transiently rebounded after 6 months. We identified two notable differences between the Pt-tropic HIV-1 and SIVmne. First, SIV Vif does not associate with Pt-tropic HIV-1 viral particles. Second, while Pt-tropic HIV-1 degrades both Pt APOBEC3G and APOBEC3F, it prevents their inclusion in virions to a lesser extent than pathogenic SIVmne. Thus, while SIV Vif is necessary for persistent infection by Pt-tropic HIV-1, improved expression and inhibition of APOBEC3 proteins may be required for robust viral replication in vivo. Additional adaptation of the virus may also be necessary to enhance viral replication. Nevertheless, our data suggest the potential for the pig-tailed macaque to be developed as an animal model of HIV-1 infection and disease.
Collapse
|
20
|
Biesinger T, White R, Yu Kimata MT, Wilson BK, Allan JS, Kimata JT. Relative replication capacity of phenotypic SIV variants during primary infections differs with route of inoculation. Retrovirology 2010; 7:88. [PMID: 20942954 PMCID: PMC2964591 DOI: 10.1186/1742-4690-7-88] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 10/13/2010] [Indexed: 01/08/2023] Open
Abstract
Background Previous studies of human and simian immunodeficiency virus (HIV and SIV) have demonstrated that adaptive mutations selected during the course of infection alter viral replicative fitness, persistence, and pathogenicity. What is unclear from those studies is the impact of transmission on the replication and pathogenicity of the founding virus population. Using the SIV-macaque model, we examined whether the route of infection would affect the establishment and replication of two SIVmne variants of distinct in vitro and in vivo biological characteristics. For these studies, we performed dual-virus inoculations of pig-tailed macaques via intrarectal or intravenous routes with SIVmneCl8, a miminally pathogenic virus, and SIVmne027, a highly pathogenic variant that replicates more robustly in CD4+ T cells. Results The data demonstrate that SIVmne027 is the dominant virus regardless of the route of infection, indicating that the capacity to replicate efficiently in CD4+ T cells is important for fitness. Interestingly, in comparison to intravenous co-infection, intrarectal inoculation enabled greater relative replication of the less pathogenic virus, SIVmneCl8. Moreover, a higher level of SIVmneCl8 replication during primary infection of the intrarectally inoculated macaques was associated with lower overall plasma viral load and slower decline in CD4+ T cells, even though SIVmne027 eventually became the dominant virus. Conclusions These results suggest that the capacity to replicate in CD4+ T cells is a significant determinant of SIV fitness and pathogenicity. Furthermore, the data also suggest that mucosal transmission may support early replication of phenotypically diverse variants, while slowing the rate of CD4+ T cell decline during the initial stages of infection.
Collapse
Affiliation(s)
- Tasha Biesinger
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
21
|
Ho O, Larsen K, Polacino P, Li Y, Anderson D, Song R, Ruprecht RM, Hu SL. Pathogenic infection of Macaca nemestrina with a CCR5-tropic subtype-C simian-human immunodeficiency virus. Retrovirology 2009; 6:65. [PMID: 19602283 PMCID: PMC2720380 DOI: 10.1186/1742-4690-6-65] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 07/14/2009] [Indexed: 12/22/2022] Open
Abstract
Background Although pig-tailed macaques (Macaca nemestrina) have been used in AIDS research for years, less is known about the early immunopathogenic events in this species, as compared to rhesus macaques (Macaca mulatta). Similarly, the events in early infection are well-characterized for simian immunodeficiency viruses (SIV), but less so for chimeric simian-human immunodeficiency viruses (SHIV), although the latter have been widely used in HIV vaccine studies. Here, we report the consequences of intrarectal infection with a CCR5-tropic clade C SHIV-1157ipd3N4 in pig-tailed macaques. Results Plasma and cell-associated virus was detectable in peripheral blood and intestinal tissues of all four pig-tailed macaques following intrarectal inoculation with SHIV-1157ipd3N4. We also observed a rapid and irreversible loss of CD4+ T cells at multiple mucosal sites, resulting in a marked decrease of CD4:CD8 T cell ratios 0.5–4 weeks after inoculation. This depletion targeted subsets of CD4+ T cells expressing the CCR5 coreceptor and having a CD28-CD95+ effector memory phenotype, consistent with the R5-tropism of SHIV-1157ipd3N4. All three animals that were studied beyond the acute phase seroconverted as early as week 4, with two developing cross-clade neutralizing antibody responses by week 24. These two animals also demonstrated persistent plasma viremia for >48 weeks. One of these animals developed AIDS, as shown by peripheral blood CD4+ T-cell depletion starting at 20 weeks post inoculation. Conclusion These findings indicate that SHIV-1157ipd3N4-induced pathogenesis in pig-tailed macaques followed a similar course as SIV-infected rhesus macaques. Thus, R5 SHIV-C-infection of pig-tailed macaques could provide a useful and relevant model for AIDS vaccine and pathogenesis research.
Collapse
Affiliation(s)
- On Ho
- Department of Pharmaceutics, University of Washington, Seattle, 98195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Polacino P, Larsen K, Galmin L, Suschak J, Kraft Z, Stamatatos L, Anderson D, Barnett SW, Pal R, Bost K, Bandivdekar AH, Miller CJ, Hu SL. Differential pathogenicity of SHIV infection in pig-tailed and rhesus macaques. J Med Primatol 2009; 37 Suppl 2:13-23. [PMID: 19187427 DOI: 10.1111/j.1600-0684.2008.00325.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Differential pathogenicity has been observed in cynomolgus and rhesus macaques following primate lentivirus infection. However, little is known about the comparative susceptibility of pig-tailed macaques to lentivirus infection and diseases. METHODS We compared the in vivo infectivity and pathogenicity of a CCR5-tropic SHIV(SF162 P4) after intravenous, intravaginal or intrarectal inoculation in rhesus and pig-tailed macaques. Plasma viral load, peripheral blood CD4(+) T cell counts and clinical signs were monitored. RESULTS Both rhesus and pig-tailed macaques are similarly susceptible to SHIV(SF162 P4) infection by intravenous and mucosal routes. However, infection was significantly more robust in pig-tailed macaques than in rhesus, resulting in persistent viremia in 9/21 pig-tails vs. 2/24 rhesus (P < 0.013) and severe CD4(+) T-cell depletion in 2/21 pig-tails (vs. none in rhesus). CONCLUSIONS Together with earlier observations, our findings underscore the importance of considering host genetic and immunological factors when comparing vaccine efficacy in different macaque species.
Collapse
Affiliation(s)
- Patricia Polacino
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|