1
|
Tisoncik-Go J, Lewis TB, Whitmore LS, Voss K, Niemeyer S, Dai J, Kim P, Hubbell K, Iwayama N, Ahrens C, Wangari S, Murnane R, Edlefsen PT, Guerriero KA, Gale M, Fuller DH, O'Connor MA. Chronic innate immune impairment and ZIKV persistence in the gastrointestinal tract during SIV infection in pigtail macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609309. [PMID: 39229223 PMCID: PMC11370579 DOI: 10.1101/2024.08.23.609309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mosquito borne flaviviruses, including dengue (DENV) and Zika (ZIKV) viruses, have caused global epidemics in areas with high HIV prevalence due to the expanded geographic range of arthropod vectors. Despite the occurrence of large flavivirus outbreaks in countries with high HIV prevalence, there is little knowledge regarding the effects of flavivirus infection in people living with HIV (PLWH). Here, we use a pigtail macaque model of HIV/AIDS to investigate the impact of simian immunodeficiency virus (SIV)-induced immunosuppression on ZIKV replication and pathogenesis. Early acute SIV infection induced expansion of peripheral ZIKV cellular targets and increased innate immune activation and peripheral blood mononuclear cells (PBMC) from SIV infected macaques were less permissive to ZIKV infection in vitro. In SIV-ZIKV co-infected animals, we found increased persistence of ZIKV in the periphery and tissues corresponding to alterations in innate cellular (monocytes, neutrophils) recruitment to the blood and tissues, decreased anti-ZIKV immunity, and chronic peripheral inflammatory and innate immune gene expression. Collectively, these findings suggest that untreated SIV infection may impair cellular innate responses and create an environment of chronic immune activation that promotes prolonged ZIKV viremia and persistence in the gastrointestinal tract. These results suggest that PLWH or other immunocompromised individuals could be at a higher risk for chronic ZIKV replication, which in turn could increase the timeframe of ZIKV transmission. Thus, PLWH are important populations to target during the deployment of vaccine and treatment strategies against ZIKV.
Collapse
Affiliation(s)
- Jennifer Tisoncik-Go
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Immunology, University of Washington (Seattle, Washington)
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington (Seattle, Washington)
| | - Thomas B Lewis
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Leanne S Whitmore
- Department of Immunology, University of Washington (Seattle, Washington)
| | - Kathleen Voss
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Immunology, University of Washington (Seattle, Washington)
| | - Skyler Niemeyer
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Jin Dai
- Department of Immunology, University of Washington (Seattle, Washington)
| | - Paul Kim
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Kai Hubbell
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Naoto Iwayama
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Chul Ahrens
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Robert Murnane
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | | | - Kathryn A Guerriero
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Michael Gale
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Immunology, University of Washington (Seattle, Washington)
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington (Seattle, Washington)
- Department of Global Health, University of Washington (Seattle, Washington)
| | - Deborah H Fuller
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Megan A O'Connor
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Microbiology, University of Washington (Seattle, Washington)
| |
Collapse
|
2
|
Carrillo MA, Zhen A, Mu W, Rezek V, Martin H, Peterson CW, Kiem HP, Kitchen SG. Stem cell-derived CAR T cells show greater persistence, trafficking, and viral control compared to ex vivo transduced CAR T cells. Mol Ther 2024; 32:1000-1015. [PMID: 38414243 PMCID: PMC11163220 DOI: 10.1016/j.ymthe.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/19/2024] [Accepted: 02/24/2024] [Indexed: 02/29/2024] Open
Abstract
Adoptive cell therapy (ACT) using T cells expressing chimeric antigen receptors (CARs) is an area of intense investigation in the treatment of malignancies and chronic viral infections. One of the limitations of ACT-based CAR therapy is the lack of in vivo persistence and maintenance of optimal cell function. Therefore, alternative strategies that increase the function and maintenance of CAR-expressing T cells are needed. In our studies using the humanized bone marrow/liver/thymus (BLT) mouse model and nonhuman primate (NHP) model of HIV infection, we evaluated two CAR-based gene therapy approaches. In the ACT approach, we used cytokine enhancement and preconditioning to generate greater persistence of anti-HIV CAR+ T cells. We observed limited persistence and expansion of anti-HIV CAR T cells, which led to minimal control of the virus. In our stem cell-based approach, we modified hematopoietic stem/progenitor cells (HSPCs) with anti-HIV CAR to generate anti-HIV CAR T cells in vivo. We observed CAR-expressing T cell expansion, which led to better plasma viral load suppression. HSPC-derived CAR cells in infected NHPs showed superior trafficking and persistence in multiple tissues. Our results suggest that a stem cell-based CAR T cell approach may be superior in generating long-term persistence and functional antiviral responses against HIV infection.
Collapse
Affiliation(s)
- Mayra A Carrillo
- Department of Medicine, Division of Hematology and Oncology, and UCLA AIDS Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Anjie Zhen
- Department of Medicine, Division of Hematology and Oncology, and UCLA AIDS Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Wenli Mu
- Department of Medicine, Division of Hematology and Oncology, and UCLA AIDS Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Valerie Rezek
- Department of Medicine, Division of Hematology and Oncology, and UCLA AIDS Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Heather Martin
- Department of Medicine, Division of Hematology and Oncology, and UCLA AIDS Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Christopher W Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Scott G Kitchen
- Department of Medicine, Division of Hematology and Oncology, and UCLA AIDS Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Broad Stem Cell Research Center, Jonsson Comprehensive Cancer Center, and Molecular Biology Institute, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
3
|
O’Connor MA, Erasmus JH, Randall S, Archer J, Lewis TB, Brown B, Fredericks M, Groenier S, Iwayama N, Ahrens C, Garrison W, Wangari S, Guerriero KA, Fuller DH. A Single Dose SARS-CoV-2 Replicon RNA Vaccine Induces Cellular and Humoral Immune Responses in Simian Immunodeficiency Virus Infected and Uninfected Pigtail Macaques. Front Immunol 2021; 12:800723. [PMID: 34992610 PMCID: PMC8724308 DOI: 10.3389/fimmu.2021.800723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
The ongoing COVID-19 vaccine rollout is critical for reducing SARS-CoV-2 infections, hospitalizations, and deaths worldwide. Unfortunately, massive disparities exist in getting vaccines to vulnerable populations, including people living with HIV. Preliminary studies indicate that COVID-19 mRNA vaccines are safe and immunogenic in people living with HIV that are virally suppressed with potent antiretroviral therapy but may be less efficacious in immunocompromised individuals. This raises the concern that COVID-19 vaccines may be less effective in resource poor settings with limited access to antiretroviral therapy. Here, we evaluated the immunogenicity of a single dose COVID-19 replicon RNA vaccine expressing Spike protein (A.1) from SARS-CoV-2 (repRNA-CoV2S) in immunocompromised, SIV infected and immune competent, naïve pigtail macaques. Moderate vaccine-specific cellular Th1 T-cell responses and binding and neutralizing antibodies were induced by repRNA-CoV2S in SIV infected animals and naïve animals. Furthermore, vaccine immunogenicity was elicited even among the animals with the highest SIV viral burden or lowest peripheral CD4 counts prior to immunization. This study provides evidence that a SARS-CoV-2 repRNA vaccine could be employed to induce strong immunity against COVID-19 in HIV infected and other immunocompromised individuals.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Cells, Cultured
- Disease Models, Animal
- Host-Pathogen Interactions
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunocompromised Host
- Immunogenicity, Vaccine
- Macaca nemestrina
- Male
- Simian Acquired Immunodeficiency Syndrome/blood
- Simian Acquired Immunodeficiency Syndrome/immunology
- Simian Acquired Immunodeficiency Syndrome/virology
- Simian Immunodeficiency Virus/immunology
- Simian Immunodeficiency Virus/pathogenicity
- Spike Glycoprotein, Coronavirus/administration & dosage
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/virology
- Time Factors
- Vaccination
- Vaccine Efficacy
- mRNA Vaccines/administration & dosage
- mRNA Vaccines/genetics
- mRNA Vaccines/immunology
Collapse
Affiliation(s)
- Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, WA, United States
- HDT Bio, Seattle, WA, United States
| | - Samantha Randall
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Jacob Archer
- Department of Microbiology, University of Washington, Seattle, WA, United States
- HDT Bio, Seattle, WA, United States
| | - Thomas B. Lewis
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Brieann Brown
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Megan Fredericks
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Skyler Groenier
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Naoto Iwayama
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Chul Ahrens
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - William Garrison
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Kathryn A. Guerriero
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| |
Collapse
|
4
|
A Gut Reaction to SIV and SHIV Infection: Lower Dysregulation of Mucosal T Cells during Acute Infection Is Associated with Greater Viral Suppression during cART. Viruses 2021; 13:v13081609. [PMID: 34452474 PMCID: PMC8402906 DOI: 10.3390/v13081609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/31/2021] [Accepted: 08/12/2021] [Indexed: 11/27/2022] Open
Abstract
Selection of a pre-clinical non-human primate (NHP) model is essential when evaluating therapeutic vaccine and treatment strategies for HIV. SIV and SHIV-infected NHPs exhibit a range of viral burdens, pathologies, and responses to combinatorial antiretroviral therapy (cART) regimens and the choice of the NHP model for AIDS could influence outcomes in studies investigating interventions. Previously, in rhesus macaques (RMs) we showed that maintenance of mucosal Th17/Treg homeostasis during SIV infection correlated with a better virological response to cART. Here, in RMs we compared viral kinetics and dysregulation of gut homeostasis, defined by T cell subset disruption, during highly pathogenic SIVΔB670 compared to SHIV-1157ipd3N4 infection. SHIV infection resulted in lower acute viremia and less disruption to gut CD4 T-cell homeostasis. Additionally, 24/24 SHIV-infected versus 10/19 SIV-infected animals had sustained viral suppression <100 copies/mL of plasma after 5 months of cART. Significantly, the more profound viral suppression during cART in a subset of SIV and all SHIV-infected RMs corresponded with less gut immune dysregulation during acute SIV/SHIV infection, defined by maintenance of the Th17/Treg ratio. These results highlight significant differences in viral control during cART and gut dysregulation in NHP AIDS models and suggest that selection of a model may impact the evaluation of candidate therapeutic interventions for HIV treatment and cure strategies.
Collapse
|
5
|
Cardozo-Ojeda EF, Duke ER, Peterson CW, Reeves DB, Mayer BT, Kiem HP, Schiffer JT. Thresholds for post-rebound SHIV control after CCR5 gene-edited autologous hematopoietic cell transplantation. eLife 2021; 10:57646. [PMID: 33432929 PMCID: PMC7803377 DOI: 10.7554/elife.57646] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 12/27/2020] [Indexed: 01/10/2023] Open
Abstract
Autologous, CCR5 gene-edited hematopoietic stem and progenitor cell (HSPC) transplantation is a promising strategy for achieving HIV remission. However, only a fraction of HSPCs can be edited ex vivo to provide protection against infection. To project the thresholds of CCR5-edition necessary for HIV remission, we developed a mathematical model that recapitulates blood T cell reconstitution and plasma simian-HIV (SHIV) dynamics from SHIV-1157ipd3N4-infected pig-tailed macaques that underwent autologous transplantation with CCR5 gene editing. The model predicts that viral control can be obtained following analytical treatment interruption (ATI) when: (1) transplanted HSPCs are at least fivefold higher than residual endogenous HSPCs after total body irradiation and (2) the fraction of protected HSPCs in the transplant achieves a threshold (76–94%) sufficient to overcome transplantation-dependent loss of SHIV immunity. Under these conditions, if ATI is withheld until transplanted gene-modified cells engraft and reconstitute to a steady state, spontaneous viral control is projected to occur.
Collapse
Affiliation(s)
- E Fabian Cardozo-Ojeda
- Vaccine and Infectious Disease Division, University of Washington, Seattle, United States
| | - Elizabeth R Duke
- Vaccine and Infectious Disease Division, University of Washington, Seattle, United States.,Department of Medicine, University of Washington, Seattle, United States
| | - Christopher W Peterson
- Department of Medicine, University of Washington, Seattle, United States.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, United States.,Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Daniel B Reeves
- Vaccine and Infectious Disease Division, University of Washington, Seattle, United States
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, University of Washington, Seattle, United States
| | - Hans-Peter Kiem
- Department of Medicine, University of Washington, Seattle, United States.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, United States.,Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, United States.,Department of Pathology, University of Washington, Seattle, United States
| | - Joshua T Schiffer
- Vaccine and Infectious Disease Division, University of Washington, Seattle, United States.,Department of Medicine, University of Washington, Seattle, United States.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, United States
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Zhen A, Peterson CW, Carrillo MA, Reddy SS, Youn CS, Lam BB, Chang NY, Martin HA, Rick JW, Kim J, Neel NC, Rezek VK, Kamata M, Chen ISY, Zack JA, Kiem HP, Kitchen SG. Long-term persistence and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman primate model of HIV/AIDS. PLoS Pathog 2017; 13:e1006753. [PMID: 29284044 PMCID: PMC5746250 DOI: 10.1371/journal.ppat.1006753] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cells have emerged as a powerful immunotherapy for various forms of cancer and show promise in treating HIV-1 infection. However, significant limitations are persistence and whether peripheral T cell-based products can respond to malignant or infected cells that may reappear months or years after treatment remains unclear. Hematopoietic Stem/Progenitor Cells (HSPCs) are capable of long-term engraftment and have the potential to overcome these limitations. Here, we report the use of a protective CD4 chimeric antigen receptor (C46CD4CAR) to redirect HSPC-derived T-cells against simian/human immunodeficiency virus (SHIV) infection in pigtail macaques. CAR-containing cells persisted for more than 2 years without any measurable toxicity and were capable of multilineage engraftment. Combination antiretroviral therapy (cART) treatment followed by cART withdrawal resulted in lower viral rebound in CAR animals relative to controls, and demonstrated an immune memory-like response. We found CAR-expressing cells in multiple lymphoid tissues, decreased tissue-associated SHIV RNA levels, and substantially higher CD4/CD8 ratios in the gut as compared to controls. These results show that HSPC-derived CAR T-cells are capable of long-term engraftment and immune surveillance. This study demonstrates for the first time the safety and feasibility of HSPC-based CAR therapy in a large animal preclinical model. Hematopoietic Stem/Progenitor Cell (HSPC) based gene therapy can be used to treat many infectious and genetic diseases. Here, we used an HSPC-based approach to redirect and enhance host immunity against HIV-1. We engineered HSPCs to carry chimeric antigen receptor (CAR) genes that detect and destroy HIV-infected cells. CAR therapy has shown huge potential in the treatment of cancer, but has only been applied in peripheral blood T-cells. HSPC-based CAR therapy has several benefits over T cell gene therapy, as it allows for normal T cell development, selection, and persistence of the engineered cells for the lifetime of the patient. We used a CAR molecule that hijacks the essential interaction between the virus and the cell surface molecule CD4 to redirect HSPC-derived T-cells against infected cells. We observed >2 years of stable production of CAR-expressing cells without any adverse events, and wide distribution of these cells in lymphoid tissues and gastrointestinal tract, which are major anatomic sites for HIV replication and persistence in suppressed patients. Most importantly, HSPC-derived CAR T-cells functionally responded to infected cells. This study demonstrates for the first time the safety and feasibility of HSPC based therapy utilizing an HIV-specific CAR for suppressed HIV infection.
Collapse
Affiliation(s)
- Anjie Zhen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Christopher W. Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Departments of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Mayra A. Carrillo
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Sowmya Somashekar Reddy
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Cindy S. Youn
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Brianna B. Lam
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Nelson Y. Chang
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Heather A. Martin
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jonathan W. Rick
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Kim
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Nick C. Neel
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Valerie K. Rezek
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Masakazu Kamata
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
| | - Irvin S. Y. Chen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, California, United States of America
| | - Jerome A. Zack
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, California, United States of America
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Departments of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Scott G. Kitchen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
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
|
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
|
17
|
Positive selection of mC46-expressing CD4+ T cells and maintenance of virus specific immunity in a primate AIDS model. Blood 2013; 122:179-87. [PMID: 23719296 DOI: 10.1182/blood-2013-01-482224] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Despite continued progress in the development of novel antiretroviral therapies, it has become increasingly evident that drug-based treatments will not lead to a functional or sterilizing cure for HIV(+) patients. In 2009, an HIV(+) patient was effectively cured of HIV following allogeneic transplantation of hematopoietic stem cells (HSCs) from a CCR5(-/-) donor. The utility of this approach, however, is severely limited because of the difficulty in finding matched donors. Hence, we studied the potential of HIV-resistant stem cells in the autologous setting in a nonhuman primate AIDS model and incorporated a fusion inhibitor (mC46) as the means for developing infection-resistant cells. Pigtail macaques underwent identical transplants and Simian-Human Immunodeficiency Virus (SHIV) challenge procedures with the only variation between control and mC46 macaques being the inclusion of a fusion-inhibitor expression cassette. Following SHIV challenge, mC46 macaques, but not control macaques, showed a positive selection of gene-modified CD4(+) T cells in peripheral blood, gastrointestinal tract, and lymph nodes, accounting for >90% of the total CD4(+) T-cell population. mC46 macaques also maintained high frequencies of SHIV-specific, gene-modified CD4(+) T cells, an increase in nonmodified CD4(+) T cells, enhanced cytotoxic T lymphocyte function, and antibody responses. These data suggest that HSC protection may be a potential alternative to conventional antiretroviral therapy in patients with HIV/AIDS.
Collapse
|
18
|
Ren W, Mumbauer A, Zhuang K, Harbison C, Knight H, Westmoreland S, Gettie A, Blanchard J, Cheng-Mayer C. Mucosal transmissibility, disease induction and coreceptor switching of R5 SHIVSF162P3N molecular clones in rhesus macaques. Retrovirology 2013; 10:9. [PMID: 23369442 PMCID: PMC3571932 DOI: 10.1186/1742-4690-10-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 01/25/2013] [Indexed: 11/18/2022] Open
Abstract
Background Mucosally transmissible and pathogenic CCR5 (R5)-tropic simian-human immunodeficiency virus (SHIV) molecular clones are useful reagents to identity neutralization escape in HIV-1 vaccine experiments and to study the envelope evolutionary process and mechanistic basis for coreceptor switch during the course of natural infection. Results We observed progression to AIDS in rhesus macaques infected intrarectally with molecular clones of the pathogenic R5 SHIVSF162P3N isolate. Expansion to CXCR4 usage was documented in one diseased macaque that mounted a neutralizing antibody response and in another that failed to do so, with the latter displaying a rapid progressor phenotype. V3 loop envelop glycoprotein gp120 sequence changes that are predictive of a CXCR4 (X4)-using phenotype in HIV-1 subtype B primary isolates, specifically basic amino acid substations at positions 11 (S11R), 24 (G24R) and 25 (D25K) of the loop were detected in the two infected macaques. Functional assays showed that envelopes with V3 S11R or D25K mutation were dual-tropic, infecting CD4+ target cells that expressed either the CCR5 or CXCR4 coreceptor. And, consistent with findings of coreceptor switching in macaques infected with the pathogenic isolate, CXCR4-using variant was first detected in the lymph node of the chronically infected rhesus monkey several weeks prior to its presence in peripheral blood. Moreover, X4 emergence in this macaque coincided with persistent peripheral CD4+ T cell loss and a decline in neutralizing antibody titer that are suggestive of immune deterioration, with macrophages as the major virus-producing cells at the end-stage of disease. Conclusions The data showed that molecular clones derived from the R5 SHIVSF162P3N isolate are mucosally transmissible and induced disease in a manner similar to that observed in HIV-1 infected individuals, providing a relevant and useful animal infection model for in-depth analyses of host selection pressures and the env evolutionary changes that influence disease outcome, coreceptor switching and vaccine escape.
Collapse
Affiliation(s)
- Wuze Ren
- Aaron Diamond AIDS Research Center, 455 First Ave,, 7th Floor, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Louz D, Bergmans HE, Loos BP, Hoeben RC. Animal models in virus research: their utility and limitations. Crit Rev Microbiol 2012; 39:325-61. [PMID: 22978742 DOI: 10.3109/1040841x.2012.711740] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Viral diseases are important threats to public health worldwide. With the number of emerging viral diseases increasing the last decades, there is a growing need for appropriate animal models for virus studies. The relevance of animal models can be limited in terms of mimicking human pathophysiology. In this review, we discuss the utility of animal models for studies of influenza A viruses, HIV and SARS-CoV in light of viral emergence, assessment of infection and transmission risks, and regulatory decision making. We address their relevance and limitations. The susceptibility, immune responses, pathogenesis, and pharmacokinetics may differ between the various animal models. These complexities may thwart translating results from animal experiments to the humans. Within these constraints, animal models are very informative for studying virus immunopathology and transmission modes and for translation of virus research into clinical benefit. Insight in the limitations of the various models may facilitate further improvements of the models.
Collapse
Affiliation(s)
- Derrick Louz
- National Institute for Public Health and the Environment (RIVM), GMO Office , Bilthoven , The Netherlands
| | | | | | | |
Collapse
|
20
|
Simian immunodeficiency virus-induced alterations in monocyte production of tumor necrosis factor alpha contribute to reduced immune activation in sooty mangabeys. J Virol 2012; 86:7605-15. [PMID: 22553338 DOI: 10.1128/jvi.06813-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is characterized by persistent viral replication in the context of CD4(+) T cell depletion and elevated immune activation associated with disease progression. In contrast, simian immunodeficiency virus (SIV) infection of African-origin sooty mangabeys (SM) generally does not result in simian AIDS despite high viral loads and therefore affords a unique model in which to study the immunologic contributions to a nonpathogenic lentiviral disease outcome. A key feature of these natural SIV infections is the maintenance of low levels of immune activation during chronic infection. Our goal was to delineate the contribution of monocytes to maintaining low levels of immune activation in SIV-infected SM. Utilizing an ex vivo whole-blood assay, proinflammatory cytokine production was quantified in monocytes in response to multiple Toll-like receptor (TLR) ligands and a specific, significant reduction in the tumor necrosis factor alpha (TNF-α) response to lipopolysaccharide (LPS) was observed in SIV-infected SM. In contrast, monocytes from hosts of pathogenic infections (HIV-infected humans and SIV-infected Asian macaques) maintained a robust TNF-α response. In SIV-infected SM, monocyte TNF-α responses to low levels of LPS could be augmented by the presence of plasma from uninfected control animals. The impact of LPS-induced TNF-α production on immune activation was demonstrated in vitro, as TNF-α blocking antibodies inhibited downstream CD8(+) T cell activation in a dose-dependent manner. These data demonstrate an association between nonpathogenic SIV infection of SM and a reduced monocyte TNF-α response to LPS, and they identify a role for monocytes in contributing to the suppressed chronic immune activation observed in these natural hosts.
Collapse
|
21
|
Tso FY, Tully DC, Gonzalez S, Quince C, Ho O, Polacino P, Ruprecht RM, Hu SL, Wood C. Dynamics of envelope evolution in clade C SHIV-infected pig-tailed macaques during disease progression analyzed by ultra-deep pyrosequencing. PLoS One 2012; 7:e32827. [PMID: 22427893 PMCID: PMC3299704 DOI: 10.1371/journal.pone.0032827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 02/06/2012] [Indexed: 11/19/2022] Open
Abstract
Understanding the evolution of the human immunodeficiency virus type 1 (HIV-1) envelope during disease progression can provide tremendous insights for vaccine development, and simian-human immunodeficiency virus (SHIV) infection of non-human primate provides an ideal platform for such studies. A newly developed clade C SHIV, SHIV-1157ipd3N4, which was able to infect rhesus macaques, closely resembled primary HIV-1 in transmission and pathogenesis, was used to infect several pig-tailed macaques. One of the infected animals subsequently progressed to AIDS, whereas one remained a non-progressor. The viral envelope evolution in the infected animals during disease progression was analyzed by a bioinformatics approach using ultra-deep pyrosequencing. Our results showed substantial envelope variations emerging in the progressor animal after the onset of AIDS. These envelope variations impacted the length of the variable loops and charges of different envelope regions. Additionally, multiple mutations were located at the CD4 and CCR5 binding sites, potentially affecting receptor binding affinity, viral fitness and they might be selected at late stages of disease. More importantly, these envelope mutations are not random since they had repeatedly been observed in a rhesus macaque and a human infant infected by either SHIV or HIV-1, respectively, carrying the parental envelope of the infectious molecular clone SHIV-1157ipd3N4. Moreover, similar mutations were also observed from other studies on different clades of envelopes regardless of the host species. These recurring mutations in different envelopes suggest that there may be a common evolutionary pattern and selection pathway for the HIV-1 envelope during disease progression.
Collapse
Affiliation(s)
- For Yue Tso
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Damien C. Tully
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Sandra Gonzalez
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Christopher Quince
- School of Engineering, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - On Ho
- Department of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Patricia Polacino
- The Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Ruth M. Ruprecht
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shiu-Lok Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
- The Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Charles Wood
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
22
|
Role of nonhuman primates in the evaluation of candidate AIDS vaccines: an industry perspective. Curr Opin HIV AIDS 2011; 5:377-85. [PMID: 20978377 DOI: 10.1097/coh.0b013e32833d2e19] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW To consider how nonhuman primate (NHP) model systems can best contribute to HIV vaccine development. RECENT FINDINGS We review the traditional roles of NHP model systems in vaccine development and compare this with how NHP models have been used in HIV vaccine research and development. Comparisons of the immune responses elicited by cellular immune response-inducing vaccines in macaques and humans illustrate the value of primate studies for the relative ranking of HIV vaccine concepts for their likely immunogenicity in humans. The unusual structures (e.g. long complementarity-determining regions) of known broadly neutralizing HIV antibodies (bNAbs) suggest that it is critical to test candidate env immunogens in NHPs, whose germline antibody repertoires resemble those of humans. Recent clinical efficacy trial results question the utility of existing NHP challenge models in predicting HIV vaccine efficacy in humans, and highlight the need to further develop models in which acquisition of infection can be reliably evaluated. When evaluated in models using low virus dose challenges that better approximate human sexual exposure to HIV - some vaccine and passive NAb interventions appear to protect against acquisition of infection. SUMMARY NHP models have important roles in the preclinical evaluation, optimization, and ranking of novel HIV immunogens. The apparent vaccine efficacy observed using low virus dose challenge models provides an opportunity to investigate the correlates of protection.
Collapse
|
23
|
Klatt NR, Harris LD, Vinton CL, Sung H, Briant JA, Tabb B, Morcock D, McGinty JW, Lifson JD, Lafont BA, Martin MA, Levine AD, Estes JD, Brenchley JM. Compromised gastrointestinal integrity in pigtail macaques is associated with increased microbial translocation, immune activation, and IL-17 production in the absence of SIV infection. Mucosal Immunol 2010; 3:387-98. [PMID: 20357762 PMCID: PMC2891168 DOI: 10.1038/mi.2010.14] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pigtail macaques (PTMs) rapidly progress to AIDS after simian immunodeficiency virus (SIV) infection. Given the strong association between human immunodeficiency virus (HIV) and SIV disease progression and microbial translocation and immune activation, we assessed whether high basal levels of immune activation and microbial translocation exist in PTMs. We found that before SIV infection, PTMs had high levels of microbial translocation that correlated with significant damage to the structural barrier of the gastrointestinal tract. Moreover, this increased microbial translocation correlated with high levels of immune activation and was associated with high frequencies of interleukin-17-producing T cells. These data highlight the relationship among mucosal damage, microbial translocation and systemic immune activation in the absence of SIV replication, and underscore the importance of microbial translocation in the rapid course of disease progression in SIV-infected PTMs. Furthermore, these data suggest that PTM may be an ideal model to study therapeutic interventions aimed at decreasing microbial translocation-induced immune activation.
Collapse
Affiliation(s)
- N R Klatt
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|